234
Assessment of missense alterations in MLH1 and their pathogenic significance by Needra Sheron Perera A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Laboratory Medicine and Pathobiology University of Toronto © Copyright by Needra Sheron Perera 2010

Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

Assessment of missense alterations in MLH1 and their pathogenic

significance

by

Needra Sheron Perera

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Department of Laboratory Medicine and Pathobiology

University of Toronto

© Copyright by Needra Sheron Perera 2010

Page 2: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

ii

Assessment of missense alterations in MLH1 and their pathogenic

significance

Needra Sheron Perera

Doctor of Philosophy

Department of Laboratory Medicine and Pathobiology

University of Toronto

2010

Abstract

Germline mutations in mismatch repair genes predispose individuals to Lynch Syndrome, the

most common colorectal cancer predisposition syndrome. MLH1 is a key mismatch repair gene

that is mutated in Lynch syndrome and about a third of the genetic alterations identified in MLH1

are missense variants of unclassified clinical significance. We hypothesize that missense

alterations in distinct domains of MLH1 likely affect its expression and function(s) to varying

degrees. To address this we utilized several approaches to investigate the molecular basis of the

pathogenicity of a panel of unclassified variants. Our results demonstrate that the MLH1 variants

p.R265C and p.K618A significantly decrease the stability of the MLH1 protein, while the variant

p.L749Q compromises heterodimerization of the MLH1-PMS2 complex. Given the limitations

and complexity of in vitro assessment strategies, we conducted a proof-of-principle study to

investigate whether missense variants in MLH1 lead to allelic imbalances at the transcriptional

level. Our analysis using the PeakPicker software indicated that the missense variants c.350C>T,

c.793C>T, c.1852_1853AA>GC, as well as the truncating variant c.1528C>T were associated

with significantly unbalanced allelic expression. This illustrates a novel method of investigating

the pathogenicity of unclassified genetic variants, which has the potential to be applied in the

Page 3: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

iii

diagnostic setting. Previous genetic epidemiology studies from our laboratory have demonstrated

that the MLH1 c.-93G>A promoter variant is strongly associated with the microsatellite

instability phenotype in colorectal tumours. Additionally, this promoter variant was associated

with an elevated risk of endometrial cancer in case-control studies. Results from our functional

studies indicate that the c.-93G>A variant significantly alters the promoter activity of MLH1.

The MLH1 promoter is bi-directional with the EPM2AIP1 gene located on the antisense strand.

Interestingly, we observed that this variant significantly affected EPM2AIP1 transcription as

well. Furthermore, our experiments suggest that c.-93G>A variant affects transcription by

altering the affinity of nuclear factors that bind this region. Combined, these findings shed light

on the possible mechanisms by which missense variants affect MLH1 expression and function,

which in conjunction with results from other functional assays will help increase the accuracy

and efficiency of genetic testing of inherited cancers.

Page 4: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

iv

Acknowledgments

First and foremost I would like to thank my supervisor and mentor, Dr. Bharati Bapat for giving

me the chance to train in her laboratory. Her intellectual guidance and advice were pivotal in

helping me succeed in my education. I also appreciate her sincere concern and encouragement in

all matters of my life.

I would like to thank all the past and present members of the Bapat laboratory, especially Vaiju

Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li,

Ted Chappell, Roula Raptis, Pinella Buongiorno, Susmita Esufali, Ekaterina Olkhov, Bas

VanRhijn, as well as Joyce Keating for their help and advice . Over the years we have

transitioned from co-workers to friends and I hope we remain so in the future.

I am grateful to my committee members, Dr. Eleftherios Diamandis and Dr. Ben Alman, for their

support and guidance over the years. A special thank you to Dr. Harry Elsholtz, whose advice

and encouragement has helped me through the various stages of my graduate education. I would

also like to thank Dr. Hilmi Ozcelik and the members of the Ozcelik laboratory for their

friendship over the years. Thank you also to all the members of the Dennis/Swallow laboratory,

especially Ken Lau and Yossr Haffani. A special thank you also to my external examiner, Dr.

Patricia Tonin for helping strengthen my thesis. I also appreciate the assistance of the members

of the Molecular Diagnostics Laboratory and the Biospecimen Repository, especially Lily

Ramyar and Soultana Tsitsikotas. I would also like to thank the genetic counselors, Kara

Semotiuk and Melyssa Aronson for their help with my project. I am deeply grateful to the

patients and their families for their generosity.

Page 5: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

v

I would like to thank my friend Carla Rosario, who has shared with me the joys and pains of

learning throughout the past eleven years. This journey has been richer because I have shared it

with you.

To my family and friends, especially my parents-in-law, Anton Joseph and Manel Joackhim for

their enthusiasm and interest in my education and their support over the years. A special note of

thanks to my grandfather, Basil Perera whose fatal battle with cancer and faith in an 11-year old

was the inspiration for this journey.

To my wonderful sister, Roshen Perera for always believing in me and reminding me that I could

deal with whatever life brought my way. Thank you for your constant love and your willingness

to help me in any and everything I do.

I am indebted to my beloved husband, Rohan Joackhim for his unfailing support and

encouragement. I know that my quest for academic achievement has had a profound impact on

your life and your sacrifices have not gone unnoticed. Thank you for everything!

I am deeply grateful to my wonderful parents, Florette and Brian Perera for giving me the

confidence to reach for my dreams. No words can express my gratitude for all the sacrifices you

have made for me. You have always been my greatest inspiration and strength.

Finally, my heartfelt thanks to God for making all this possible.

Page 6: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

vi

Table of Contents

ACKNOWLEDGMENTS .......................................................................................................... IV

TABLE OF CONTENTS ........................................................................................................... VI

LIST OF TABLES ....................................................................................................................... X

LIST OF FIGURES .................................................................................................................... XI

LIST OF ABBREVIATIONS ................................................................................................. XIII

CHAPTER 1 GENERAL INTRODUCTION ............................................................................ 1

1.1 COLORECTAL CANCER ...................................................................................................... 1

1.1.1 Anatomy and physiology of the Colorectum .............................................................. 1

1.1.2 Natural history and epidemiology of CRC ................................................................. 2

1.1.3 Screening, staging, and prognosis ............................................................................... 3

1.1.4 Molecular pathways in CRC ..................................................................................... 10

1.1.5 Behavioural and lifestyle risk factors ....................................................................... 26

1.1.6 Hereditary syndromes, pathology and presentation .................................................. 28

1.2 LYNCH SYNDROME ........................................................................................................... 31

1.2.1 Overview ................................................................................................................... 31

1.2.2 Pathology, presentation and clinical criteria ............................................................. 32

1.2.3 Tumour spectrum ...................................................................................................... 35

1.2.4 Predictive genetic testing .......................................................................................... 36

1.3 MISMATCH REPAIR .......................................................................................................... 37

1.3.1 Overview of the mismatch repair system in prokaryotes.......................................... 38

1.3.2 Overview of the mismatch repair system in eukaryotes ........................................... 39

1.3.3 The post-replicative error repair function ................................................................. 42

1.3.4 The role in signaling DNA damage .......................................................................... 46

1.3.5 Other roles of mismatch repair proteins .................................................................... 50

1.3.6 The structure and expression of the MMR protein MLH1 ....................................... 52

1.3.7 Mismatch repair and disease ..................................................................................... 54

1.4 GENETIC VARIATION IN MISMATCH REPAIR .................................................................. 56

Page 7: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

vii

1.4.1 Predictive models of germline variants in MMR genes............................................ 56

1.4.2 Spectrum of genetic alterations in the major MMR genes ....................................... 58

1.4.3 Approaches to classifying unclassified variants ....................................................... 60

1.5 HYPOTHESIS AND OBJECTIVES ........................................................................................ 62

CHAPTER 2 THE ROLE OF UNCLASSIFIED VARIANTS IN INFLUENCING MLH1

EXPRESSION, STABILITY AND FUNCTION ..................................................................... 64

2.1 SUMMARY .......................................................................................................................... 64

2.2 INTRODUCTION .................................................................................................................. 64

2.3 MATERIALS AND METHODS ............................................................................................... 69

2.3.1 Materials and cell culture .......................................................................................... 69

2.3.2 Identification of MLH1 variants ............................................................................... 69

2.3.3 Computational Assessment of Variants .................................................................... 70

2.3.4 Site-directed mutagenesis and construction of MLH1 mutants ................................ 70

2.3.5 RNA extraction, cDNA synthesis and sequencing ................................................... 71

2.3.6 Production of recombinant proteins and western blotting ........................................ 71

2.3.7 Combined coimmunoprecipitation and western blot analysis .................................. 72

2.3.8 Pulse chase assays ..................................................................................................... 72

2.3.9 Cycloheximide experiments...................................................................................... 73

2.3.10 Generation of stable inducible cell lines ............................................................... 73

2.3.11 Cell survival assays ............................................................................................... 75

2.3.12 Cell cycle analysis................................................................................................. 76

2.3.13 MSI analysis.......................................................................................................... 76

2.4 RESULTS ............................................................................................................................ 78

2.4.1 In silico analysis of MLH1 variants .......................................................................... 78

2.4.2 HCT116 cells are defective in MLH1 ....................................................................... 82

2.4.3 R265C and K618A decrease expression of MLH1................................................... 82

2.4.4 Altered expression is due to decreased stability of the protein ................................. 83

2.4.5 L749Q impairs ability of MLH1 to heterodimerize with PMS2............................... 88

2.4.6 HCT116 cells express inducible wild-type and variant MLH1 ................................ 90

2.4.7 Stable inducible cell lines and cell survival .............................................................. 95

2.4.8 Stable inducible cell lines and the G2/M checkpoint ................................................ 97

Page 8: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

viii

2.4.9 Stable inducible expression of MLH1 & the microsatellite instability phenotype of

the HCT116 cell line ........................................................................................................... 101

2.5 DISCUSSION ..................................................................................................................... 104

CHAPTER 3 A NOVEL AND RAPID METHOD OF DETERMINING THE EFFECT OF

MLH1 UNCLASSIFIED GENETIC VARIANTS ON DIFFERENTIAL ALLELIC

EXPRESSION ........................................................................................................................... 112

3.1 SUMMARY ........................................................................................................................ 112

3.2 INTRODUCTION ................................................................................................................ 113

3.3 MATERIALS & METHODS ................................................................................................. 115

3.3.1 Selection of Patients and Families .......................................................................... 115

3.3.2 Overview of diagnostic strategy and identification of MLH1 variants .................. 116

3.3.3 Microsatellite instability and immunohistochemistry ............................................. 116

3.3.4 DNA extraction and mutation detection ................................................................. 117

3.3.5 RNA extraction and cDNA synthesis ..................................................................... 117

3.3.6 PeakPicker Analysis................................................................................................ 117

3.3.7 Allele specific (Asymmetric) PCR ......................................................................... 119

3.3.8 In silico analysis of potential effects on splicing .................................................... 119

3.4 RESULTS .......................................................................................................................... 121

3.4.1 The truncating variant c. 1528C>T leads to unbalanced allelic expression ........... 121

3.4.2 The missense variants c. 350C>T, C. 793C>T and c.1852_1853AA>GC lead to

unbalanced allelic expression ............................................................................................. 122

3.4.3 The missense variants c. 55A>T and c. 2246T>C do not lead to unbalanced allelic

expression ........................................................................................................................... 123

3.5 DISCUSSION ..................................................................................................................... 128

CHAPTER 4 FUNCTIONAL EFFECTS OF THE MLH1-93G>A POLYMORPHISM ON

MLH1 AND EPM2AIP1 PROMOTER ACTIVITY ............................................................. 140

4.1 SUMMARY ........................................................................................................................ 140

4.2 INTRODUCTION ................................................................................................................ 141

4.3 MATERIALS AND METHODS ............................................................................................. 143

4.3.1 Generation of Promoter Constructs ........................................................................ 143

4.3.2 Cell Culture ............................................................................................................. 146

Page 9: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

ix

4.3.3 Luciferase Reporter Gene Assays ........................................................................... 146

4.3.4 Western blotting ...................................................................................................... 147

4.3.5 Electrophoretic Mobility Shift Assays (EMSA) ..................................................... 147

4.4 RESULTS .......................................................................................................................... 148

4.4.1 Effects of the MLH1 -93G>A variant on promoter activity in colorectal cancer cell

lines 148

4.4.2 Effects of the MLH1 -93G>A variant on promoter activity in endometrial cancer cell

lines 149

4.4.3 Characterization of MLH1 and EPM2AIP1 in a panel of human cell lines ........... 149

4.4.4 Effects of the MLH1 -93G>A variant on EPM2AIP1 promoter activity ............... 150

4.4.5 Effect of the MLH1 -93G>A promoter variant on transcription factor binding ..... 159

4.5 DISCUSSION ..................................................................................................................... 164

CHAPTER 5 DISCUSSION AND FUTURE DIRECTIONS ............................................... 170

REFERENCES .......................................................................................................................... 181

Page 10: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

x

List of Tables

CHAPTER 1

Table 1.1 Screening tests used to detect CRC ................................................................................ 6

Table 1.2 Tumour Stage Definitions According to the American Joint Committee on Cancer

(AJCC) ............................................................................................................................................ 8

Table 1.3 AJCC/UICC Stage Groupings ........................................................................................ 9

Table 1.4 Summary of clinical criteria used to identify Lynch syndrome.................................... 34

CHAPTER 2

Table 2.1 Primers used for site-directed mutagenesis .................................................................. 77

Table 2.2 Summary of predictions generated by computational tools .......................................... 77

Table 2.3 Summary of cell survival in response to temozolomide ............................................... 96

Table 2.4 Summary of MSI experiments .................................................................................... 102

CHAPTER 3

Table 3.1 Primers used for PeakPicker analysis ......................................................................... 120

Table 3.2 Summary of allele ratios generated by PeakPicker .................................................... 125

Table 3.3 Summary of clinicopathological characteristics ......................................................... 126

Table 3.4 Predicted effects of missense variants on splicing...................................................... 127

CHAPTER 4

Table 4.1 Summary of promoter activities ................................................................................. 158

Page 11: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xi

List of Figures

CHAPTER 1

Figure 1.1 The simplified progression of CRC via the CIN, MSI and CIMP pathways. ............. 24

Figure 1. 2 Heterodimers formed by the MutL homologs ............................................................ 40

Figure 1.3 Simplified overview of the eukaryotic MMR system. ................................................ 44

Figure 1.4 A simplified model of the MMR protein dependent DNA damage signaling network

....................................................................................................................................................... 49

CHAPTER 2

Figure 2.1 Schematic representation of the functional domains identified in MLH1. ................. 68

Figure 2.2 A Ribbon diagram of the approximate position of human MLH1 R265 on the crystal

structure of E. coli MutL. .............................................................................................................. 80

Figure 2.3 Profile of time dependent expression of wild type MLH1 in HCT116 cells. .............. 81

Figure 2.4 Expression of wild-type and variant MLH1 in HCT116 cells. ................................... 84

Figure 2.5 Expression profile of the MLH1 variants K618A & L749Q. ...................................... 85

Figure 2.6 Pulse chase assays to determine half-life of wild-type and variant protein. .............. 86

Figure 2.7 Stability of MLH1 variants as assessed by cycloheximide treatments. ....................... 87

Figure 2.8 Ability of wild-type and variant MLH1 to interact with endogenous PMS2. ............. 89

Figure 2.9 Screening of clones...................................................................................................... 92

Figure 2.10 Generation of the I19F MLH1 stable inducible cell line. .......................................... 93

Figure 2.11 Characterization of the K618A stable inducible cell line. ......................................... 94

Figure 2.12 Western blot to confirm levels of MLH1 in cell survival experiments. .................... 96

Page 12: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xii

Figure 2.13 Representative cell cycle profiles of the stable inducible and HCT116+Chr3 cell

lines in response to treatment with temozolomide. ..................................................................... 100

Figure 2.14 Expression of MLH1 and PMS2 in wild-type MLH1 cells stable inducible cells. 103

CHAPTER 3

Figure 3.1 Representative chromatograms generated by high sensitivity sequencing. .............. 124

Figure 3.2 Pedigree of Case 5A/5B. ........................................................................................... 132

Figure 3.3 Pedigree of case 6. ..................................................................................................... 133

CHAPTER 4

Figure 4.1 The schematic of the MLH1/EPM2AIP1 bi-directional promoter. ........................... 145

Figure 4.2 Effect of the MLH1-93G>A polymorphism on MLH1 transcriptional activity in the

HCT116 cell line. ........................................................................................................................ 152

Figure 4.3 Effect of the MLH1-93G>A polymorphism on MLH1 transcriptional activity in the

endometrial cancer cell lines. ...................................................................................................... 154

Figure 4.4 Expression profile of endogenous MLH1 and EPM2AIP1 in a panel of human cell

lines. ............................................................................................................................................ 155

Figure 4.5 Effect of the MLH1-93G>A polymorphism on EPM2AIP1 transcriptional activity. 157

Figure 4.6 Impact of the MLH1 -93 >A polymorphism on nuclear factor binding in HCT116. 160

Figure 4.7 Impact of the MLH1 -93 >A polymorphism on nuclear factor binding in HEC-1-A.

..................................................................................................................................................... 161

Figure 4.8 Determination of putative factors that bind the promoter region surrounding -93G>A.

..................................................................................................................................................... 163

Page 13: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xiii

List of Abbreviations

5-FU

A

ACF

AID

ALT

APC

ATM

ATR

5-Fluorouracil

Adenine

Aberrant Crypt Focus

Activation-Induced Deaminase

Alternative Lengthening of Telomeres

Adenomatous Polyposis Coli

Ataxia Telangiectasia Mutated

ATM and Rad3-related

BAC

bp

BER

BRRS

C

CS

CIMP

CIN

CNV

CRC

CXH

Bacterial Artificial Chromosome

base pair

Base Excision Repair

Bannayan-Riley-Ruvalcaba Syndrome

Cytosine

Cowden Syndrome

CpG Island Methylator Phenotype

Chromosomal Instability

Copy Number Variation

Colorectal Cancer

Cycloheximide

Page 14: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xiv

Dox

DMSO

DNA

DNTP

Doxycycline

Dimethyl Sulfoxide

Deoxyribonucleic Acid

deoxyribonucleoside triphosphate

EC

EMSA

EPM2AIP1

ES

EV

Endometrial Cancer

Electrophoretic Mobility Shift Assay

Laforin (EPM2A) Interacting Protein 1

Embryonic stem

Empty vector

Exo1

FBS

FAP

FOBT

G

hr

Exonuclease 1

Fetal Bovine Serum

Familial Adenomatous Polyposis

Fecal Occult Blood Tests

Guanine

hour

hMLH1 Human MutL Homolog 1

hMLH3 Human MutL Homolog 3

hMSH2 Human MutS Homolog 2

hMSH3 Human MutS Homolog 3

HNPCC Hereditary Nonpolyposis Colorectal Cancer

Page 15: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xv

hPMS1 Human Post-Meiotic Segregation 1

hPMS2

IDL

Human Post-Meiotic Segregation 2

Insertion Deletion Loop

IHC Immunohistochemistry

IP

JPS

Immunoprecipitate

Juvenile Polyposis Syndrome

kDa

LC

MAP

MGMT

MIM

min

miR

MLPA

kiloDalton

Lynch Syndrome

MYH-Associated Polyposis

O6-Methylguanine-DNA Methyl Transferase

Mendelian Inheritance in Man

minute

microRNA

Multiplex Ligation-dependent Probe Amplification

MMR Mismatch Repair

MNNG N-Methyl-N '-Nitro-N-Nitrosoguanidine

MNU N-Methyl-N-Nitrosourea

MSI

MSI-H

MSI-L

Microsatellite Instability

High Frequency Microsatellite Instability

Low Frequency Microsatellite Instability

Page 16: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xvi

MSS Microsatellite Stable

MutLα MLH1-PMS2 Complex

MutLβ MLH1-PMS1 Complex

MutLγ MLH1-MLH3complex

MutSα MSH2-MSH6 Complex

MutSβ

MYH

MSH2-MSH3 Complex

MutY Homolog

NLS Nuclear Localization Sequence

O6

MeG O6

Methylguanine

PCNA

PCR

Proliferating Cell Nuclear Antigen

Polymerase Chain Reaction

PI

PJS

Propidium Iodide

Peutz-Jeghers Syndrome

PolD Polymerase Delta

PolyPhen

PREMM1,2

Polymorphism Phenotyping

Prediction of Mutations in MLH1 and MSH2

RPA

RT-PCR

Replication Protein A

Reverse Transcribed Polymerase Chain Reaction

Sf9 Spodoptera frugiperda 9

SIFT Sorting Intolerant From Tolerant

Page 17: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

xvii

T Thymine

TetR

TNM

Tetracycline Repressor

Tumour-Node-Metastasis

T-rex

TMZ

V

Tetracycline-Regulated Expression system

Temozolomide

Variable region of IgM antibodies

WCL Whole Cell Lysate

wt Wild-type

Page 18: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

1

CHAPTER 1

General Introduction

1.1 Colorectal Cancer

1.1.1 Anatomy and physiology of the Colorectum

The colon is a component of the digestive system and in healthy individuals it functions to

absorb water, sodium, and chloride, while secreting potassium and bicarbonate. The components

of the colon are located either in the abdominal cavity or behind it in the retroperitoneum.

Proximal and distal segments of the large intestine (defined as regions proximal and distal to the

splenic flexure) have different embryological origins and biological characteristics [1]. The

caecum, ascending colon and proximal two-thirds of the transverse colon derive from the

midgut, while the segment comprising the splenic flexure to the upper anal canal derives from

the hindgut. The distinct embryological origins of both segments are reflected in the dual blood

supply and distinct innervations patterns. Furthermore, the pH of the lumen is lower in the

proximal colon compared to the distal colon, a factor that can affect the growth of microbes and

the metabolism of bile acids at these sites [2].

Intestinal epithelial stem cells are thought to be located at the bottom of the crypt in the colon

[3]. Their differentiating progeny are thought to migrate upwards through the transit amplifying

zone in the lower to middle region of the crypt, before becoming terminally differentiated and

eventually shed into the lumen, a process that has been estimated to take about seven days [3].

Given this continuous loss of cells, the epithelium must replace cells at an equal rate to maintain

its integrity. Thus, the intestinal epithelium has a large number of cycling cells and is one the

most rapidly renewing tissues in the body [3].

Page 19: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

2

1.1.2 Natural history and epidemiology of CRC

The multi-step genetic model of CRC carcinogenesis was first described in 1990 by Fearon and

Vogelstein[4]. According to this model the adenomatous polyposis coli (APC) tumour suppressor

gene is initially inactivated, followed by activating mutations in KRAS oncogene [4]. Subsequent

mutations in TGF-β, PIK3CA, and p53 (TP53) then drive the subsequent steps in the process of

malignant transformation [4-8]. Since then several refinements have been made to this original

model, including the inclusion of many more genes. However, this model has helped establish

several key principals that have guided our understanding and the study of CRC, as well as other

solid tumours. First, multiple genetic events are required for the malignant transformation of

cells; second, there exist several discrete intermediates in this process; and third, the temporal

acquisition of these genetic changes is an important factor that determines its outcome [9].

The earliest identifiable lesion in the colonic mucosa is the aberrant crypt focus (ACF)[10],

which has been proposed to be a putative precursor to adenocarcinoma of the colon [11]. An

ACF refers to a cluster of crypts that are characterized based on several histological features such

as darker staining, raised appearance and larger crypt size when compared to the adjacent normal

mucosa [12, 13]. The true neoplastic potential of this lesion is still unclear, but it does appear that

some of these lesions harbor mutations in KRAS or APC and can progress to adenocarcinoma

[14]. While ACF has been shown to be an important predictor for CRC, there remains a need to

further study these lesions and the context of their contribution to cancer [15, 16].

Worldwide about one million new CRC cases are diagnosed each year[17]. However, it is

predominantly a disease that affects developed nations and is the second leading cause of cancer-

related deaths in North America, affecting approximately 5% of the general population [17, 18].

It was estimated that in 2009 CRC will lead to 147,000 newly diagnosed cases as well as nearly

Page 20: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

3

50,000 deaths in the United States[19]. In Canada the estimated incidence of CRC for 2010 is

approximately 22,500 cases with approximately 9,100 deaths resulting from this disease [20].

CRC accounts for 13% of all cancers and is the third most common cancer in Canada [20]. The

incidence and mortality of CRC within Canada varies by geographic location, with the Atlantic

Provinces having the highest rates in the country and the Pacific provinces having the lowest

rates. The province of Newfoundland and Labrador has approximately twice the mortality rate of

CRC as the province of British Columbia[20]. In Ontario, approximately 8,100 CRC cases (4500

cases in men and 3800 cases in women) will be diagnosed in 2010[20]. The lifetime probability

of developing CRC is about 6.5% and the estimated five-year relative survival rate from 2002-

2004 was about 62% [21]. CRC is predominantly a disease of the elderly and more than half of

all CRCs will occur in individuals over the age of 70 [20]. Furthermore, statistical data indicate

only a slight difference between the incidence of CRC among males and females [20].

1.1.3 Screening, staging, and prognosis

The screening of asymptomatic persons, at an average risk of developing CRC allows for the

detection of cancer at early and curable stages, consequently resulting in reduced mortality [22,

23]. Furthermore, certain screening strategies are able to detect precancerous lesions, the

removal of which will result in decreased incidence of CRC [24, 25]. Currently there exist

several screening strategies each with associated strengths and limitations (Table 1.1). Fecal

occult blood tests (FOBT) can detect occult blood in small stool samples and is a non-invasive

procedure that can be performed at home. The test detects the peroxidase activity of heme and is

therefore not specific for human blood. The fecal immunohistochemical test uses antibodies

against human hemoglobin, albumin, as well as other blood components that are more specific

for human blood. However, studies indicate considerable variation in the performance of

Page 21: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

4

difference tests and the optimal commercial test is currently unknown[26]. Clinical trials have

demonstrated that those with positive occult blood tests have a three to four times higher risk of

developing cancer compared to those with negative test results[25]. Thus, colonoscopy is

recommended for those with positive test results. Furthermore, randomized control studies have

indicated that when administered annually or biennially the FOBT test results in the detection of

cancers at an earlier stage, which over a period of 10-13 years results in a reduction of mortality

from CRC of 15-33% [27-29]. Structural examinations of the colon are another type of screening

method used in CRC and computed tomographic (CT) colonography is an example of this

method. CT colonography generates two and three dimensional images of the colon, however it

is less sensitive and specific for polyps less than 6mm in diameter[25]. Barium enema

examination is another radiographic method that is able to accurately identify late-stage cancers;

however, has poor sensitivity for precursor lesions[30]. Sigmoidoscopy, another method of

structural examination cannot detect proximal lesions. This is important as it has been shown that

30% of patients with advanced CRC have only proximal lesions [31, 32]. Thus, colonoscopy is

often the preferred method as it allows for a full colon examination with polypectomy [25].

Colonoscopy is the final assessment step in any screening program and has a high level of

sensitivity. However, to date there have been no randomized, controlled trials carried out to

compare colonoscopy with other screening methods. Furthermore, it is likely that some lesions

are missed during this procedure, which is especially an issue for lesions larger than 10mm in

diameter, as well as flat adenomas [33-35]. Thus, these issues highlight the importance of

improving the quality of the colonoscopy. The recommended screening guidelines vary between

nations. In Canada, the Canadian Association of Gastroenterology and the Canadian Digestive

Health Foundation does not recommend screening for asymptomatic individuals under the age of

50 who do not have a family history of colon cancer [36]. For asymptomatic individuals over the

Page 22: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

5

age of 50 and who do not have a family history of CRC the recommendations are: 1) FOBT test

every two years, 2) a flexible sigmoidoscopy every five years, 3) flexible sigmoidoscopy

combined with FOBT every five years, 4) double contrast barium enema every five years or 5)

colonoscopy every ten years[36]. Individuals who have a family history of colon cancer have a

higher risk of disease, thus a more stringent and focused screening regimen is recommended. The

recommendations for these individuals vary depending on if they are positive for the hereditary

syndromes, as well as the number, age and closeness (first-degree vs. second-degree) of relatives

who have developed cancer.

Page 23: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

6

Table 1.1 Screening tests used to detect CRC

Test Sensitivity Advantages Limitations

Standard fecal occult

blood test

Cancer: 33-50%

AA: 11%

Ref:[27-29, 37, 38]

- Low initial cost

- Can be performed at

home

- Not specific for human

hemoglobin

- As specificity low, repeat

annual testing recommended

Sensitive fecal occult

blood test (Hemoccult

SENSA, Beckman

Coulter)

Cancer: 50-75%

AA: 20-25%

Ref: [22, 23, 38, 39]

- Low initial cost

- Can be performed at

home

- Not specific for human

hemoglobin

- As specificity low, repeat

annual testing recommended

Fecal

Immunochemical test

Cancer: 60-85%

AA: 20-50%[22, 23]

Ref: [37]

- Specific for human

hemoglobin

- Low initial cost

- Can be performed at

home

- Uncertain benefit

- Ideal number of stool

samples and repeat testing

regimen unknown

Stool DNA Cancer: 51-80%

AA: 18-40%

Ref: [39, 40]

-Detection of specific

mutations may be more

accurate

- Can be performed at

home

- Costly

- Limited to the screening of

genes known to be mutated

in CRC

- Performance of new tests

undetermined

CT colonography Cancer: Uncertain,

estimated to be

>90%

AA: estimated

>90%

Ref:[41]

- High sensitivity for

detection of lesions ≥ 10

mm in diameter

- Less invasive than

endoscopy

- Detection of polyps <6 mm

in diameter and flat polyps

uncertain

- Requires bowel preparation

and special resources

- Radiation exposure

Sigmoidoscopy Cancer: >95% in

distal colon

AA: 70%*

Ref: [31, 42]

- Office based and does

not require sedation

- Case-control studies

show 60% reduction of

mortality from cancers in

distal colon

- Does not detect proximal

colon cancer

Colonoscopy Cancer: >95%

AA: 88-98%

Ref: [31-34, 43-45]

- Case-control studies

indicate a 53-72%

reduction in

incidence[46] and 31%

reduction of

mortality[47]

- Can detect and remove

lesions during one

examination

- High initial cost

- Requires bowel preparation

and special resources

- Invasive

- 3-5 adverse reactions per

1000 examinations

Modified from Lieberman, DA[25]. AA: Advanced adenoma, a tubular adenoma that is 10 mm or larger

in diameter/an adenoma with villous histologic features/high-grade dysplasia.* If an adenoma is

detected in the distal colon the patient would undergo complete colonoscopy, which would result

in detection (if any) of adenomas in both the proximal or distal colon. Ref: References that support

these figures.

Page 24: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

7

The stage of disease in CRC determines treatment options and is the strongest predictor for

survival of CRC patients. The tumour-node-metastasis (TNM) staging system was developed by

the American Joint Committee on Cancer (AJCC) and the International Union Against Cancer

(UICC) to standardize staging for the uniform evaluation of data across specialties and nations

[48]. In this system the designation „T refers to the local extent of the untreated primary tumour

at the time of diagnosis and initial workup; „N‟ refers to the status of

the regional lymph nodes,

and „M‟ refers to distant metastatic disease (refer to Table 1.2 and Table 1.3). Pathologic

classification is based on gross/microscopic examination of the resection specimen of a

previously untreated primary tumour and clinical classification is based on evidence

acquired

through a variety of techniques that include physical examination, radiologic imaging,

endoscopy, biopsy, and surgical exploration [49]. The Dukes classification system relies on the

pathological classification of disease stages and is thus not as useful to clinicians as it cannot

be used to preoperatively evaluate the patient [49]. Despite significant progress in our

understanding over the past twenty years the 5-year survival rate for CRC remains at almost 90%

for stage I cancers to 5%–15% for stage IV disease [50, 51]. Another important feature of CRCs

are their histological grade. Tumours are graded into three categories based on the degree of

gland-like structures and thus its resemblance to the original tissue. Accordingly, Grade 1

tumours are well-differentiated, Grade 2 tumours are moderately differentiated, Grade 3 tumours

are poorly differentiated and Grade 4 tumours are undifferentiated [52]. CRC prognosis

correlates well with both stage and grade [53]. Furthermore, tumour grade was also found to

correlate significantly with the TNM staging system [52].

Page 25: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

8

Table 1.2 Tumour Stage Definitions According to the American Joint Committee on

Cancer (AJCC)

TNM Stage Description

TX

Primary tumour cannot be assessed

T0 No evidence of primary tumour

Tis Carcinoma in situ

T1 Tumour invades submucosa

T2 Tumour invades the muscularis propria

T3 Tumour invades through muscularis propria

T4 Tumour invades other organs/peritoneum

NX Regional lymph node metastasis cannot be assessed

N0 No regional lymph node metastasis

N1 Metastasis in 1- 3 regional lymph nodes

N2 Metastasis in ≥4 regional lymph nodes

MX Distant metastasis cannot be assessed

M0 No distant metastasis

M1 Distant metastasis

Page 26: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

9

Table 1.3 AJCC/UICC Stage Groupings

Stage T N M Dukes

Stage 0

Tis N0 M0 NA

Stage I

T1

T2

N0

N0

M0

M0

A

A

Stage IIA

T3 N0 M0 B

Stage IIB

T4 N0 M0 B

Stage IIIA

T1, T2 N1 M0 C

Stage IIIB

T3, T4 N1 M0 C

Stage IIIC

Any T N2 M0 C

Stage IV

Any T Any N M1 NA

Dukes: corresponding Dukes stage, NA: Not applicable

Page 27: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

10

1.1.4 Molecular pathways in CRC

CRC is a complex and heterogeneous disease with multiple genes in diverse pathways involved

in its initiation, progression, invasion and metastasis. It is widely accepted that the sequential

accumulation of mutations that activate oncogenes and disrupt tumour suppressor genes,

combined with multiple cycles of clonal selection and evolution facilitate the process of

carcinogenesis. While previous studies estimate that the disruption of about six to seven cellular

processes are required for transformation[54], a recent comprehensive sequence evaluation of

colon cancer genomes hints that this number may be even higher (about twelve) than previously

estimated [55]. Baseline mutation rates, which are estimated to be about 10-9

[56], cannot

account for the number of mutations that are required for malignant transformation. Thus, the

rate-limiting step in the process of carcinogenesis is the rate at which new mutations occur, and it

was proposed that cancer cells must acquire some form of genetic instability to facilitate the

process of carcinogenesis [57, 58]. Genetic instability refers to a set of events capable of causing

unscheduled alterations, either of a temporary or permanent nature within the genome. In CRC at

least 3 major pathways that contribute to genomic instability have been described, the

chromosomal instability (CIN) pathway, microsatellite (MSI) pathway, and CpG island

methylator (CIMP) pathway. These pathways are not mutually exclusive; therefore a tumour

may occasionally exhibit features of multiple pathways. For instance, 25% of MSI CRCs can

exhibit chromosomal instabilities [59], and about 12% of CIN-positive tumours show a high

level of MSI [60]. Furthermore, while CIMP accounts for the majority of MSI-positive/ CIN-

negative CRCs, as much as 33% of CIMP-positive tumours can exhibit a high level of

Page 28: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

11

chromosomal abnormalities [61]. Nevertheless, the biological significance of these overlapping

features are not fully understood [9].

1.1.4.1 Chromosomal instability (CIN)

CIN refers to the existence of accelerated rate of chromosomal alterations, which result in gains

or losses of whole chromosomes as well as inversions, deletions, duplications and translocations

of large chromosomal segments, which results in karyotypic variability from cell to cell [62].

CIN is considered to promote carcinogenesis through loss of tumour suppressors and the copy

numbers gains of oncogenes [63], and CIN is observed in 65-70% of CRC. Aneuploidy refers to

an abnormal karyotype and Lengauer et al have shown that aneuploidy is the result of an

abnormally high rate of CIN that persists throughout the life of the tumour [62]. The APC gene

located on chromosome 5q21 is commonly mutated in CRC. Furthermore, APC mutations are

frequently found in dysplastic aberrant crypt foci (ACF) that show the highest potential for

progressing to CRC[64]. There are two possible mechanisms by which APC mutations might

contribute to the progression of CRC, the first is by over activation of the Wingless/Wnt

signaling pathway and secondly by the induction of chromosome instability[64]. Wingless/Wnt

signaling pathway is shown to be important for several process including organ development,

cellular proliferation, morphology, motility and cell fate determination[65]. In this pathway,

APC binds to a complex of proteins that includes β-catenin and glycogen synthase kinase-3β on

an axin scaffold to form the β-catenin destruction complex (Figure 1.). This facilitates the

phosphorylation of β-catenin by glycogen synthase kinase-3β, which leads to its degradation via

the proteasome. However, in the presence of the Wnt ligand, β-catenin is no longer

phosphorylated and is thus allowed to accumulate in the cytoplasm [66, 67]. β-catenin then

translocates to the nucleus, and together with the members of T-cell factor(Tcf) and lymphoid

enhancer factor(Lef) family of transcription factors, drive the transcription of multiple genes.

Page 29: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

12

Mutations in β-catenin (CTNNB1) or APC, both of which occur in a large subset of CRCs,

increase the transcriptional activity of β-catenin [68, 69]. In the second mechanism APC leads to

chromosomal instability through its interactions with EB1. APC has an EB1-binding domain in

its C-terminal end. EB1 associates with the growing ends of microtubules and centrosomes and

has been shown to be important in the cytokinesis checkpoint in yeast [70]. APC accumulates at

the kinetochore during mitosis and Apc mutant cells form mitotic spindles with an abundance of

microtubules that are not able to connect efficiently with kinetochores [71]. Mouse embryonic

stem cells homozygous for the mutant Min (multiple intestinal neoplasia) or Apc1638T alleles

demonstrate CIN and polyploidy [71]. Although these studies implicate APC mutations in the

generation of genomic instability observed in colon adenomas and CRC, the significance of these

observations is still not clear[64]. Thus, it has been proposed that the inactivation of APC

creates a permissive state that allows the evolving tumour to tolerate the development of

aneuploidy and CIN [64, 72, 73].

The observation that cancer cells harbour an abnormal number of chromosomes was made

almost a century ago [74, 75], despite this our understanding of what causes this phenomenon

remains incomplete. To date several pathways and processes have been implicated in CIN

including; a) pathways that ensure accurate chromosome segregation, b) abnormal centrosome

number and function, c) pathways involved in telomere stability, and d) cell cycle checkpoint

pathways and kinases [9, 76]. Several of these processes will be briefly discussed below.

The mitotic checkpoint is the major cell cycle control mechanism that ensures high fidelity of

chromosome segregation. It is responsible for delaying anaphase until all pairs of duplicated

chromatids are properly aligned on the metaphase plate. Defects in this process lead to the mis-

segregation and the unequal and abnormal distribution of chromosomes into daughter cells

Page 30: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

13

resulting in aneuploidy. The mad and bub genes, two highly conserved key components of the

mitotic spindle checkpoint that appear to operate as checkpoint sensors and signal transducers

were found to be deregulated in a subset of colon cancers[77]. MAD and BUB inhibit the

anaphase-promoting complex/C (APC/C), an ubiquitin protein ligase, and also lead to cell-cycle

arrest. At anaphase, APC/C normally becomes active and ubiquitinates securin, which is then

degraded through the ubiquitin-proteasome pathway [78-80]. Securin functions by interacting with

separins, a class of caspase-related proteases that regulate cohesins, multi-protein complexes that

create physical links that maintain sister chromatids together during metaphase [81, 82]. Other

than MAD and BUB, there are additional factors that play import roles in this process. For

example, the securin-separin complex is regulated by an autofeedback loop that involves other

proteins such as polo kinases (PLK) [83]. Mutations in two genes that control the human mitotic

checkpoint, BUB1 and BUBR1(MAD3), were found in a subset of CIN colon cancer cell lines

[84]. Other genes involved in kinetochore function are reported to be either mutated or

overexpressed in colon cancer [85].

Another mechanism proposed to cause CIN is abnormal centrosome number and function. There

is a strong association between abnormal centrosome number and aneuploidy in cancers [86, 87]

The centrosome plays a central role in chromosome segregation during mitosis by serving as an

anchor for the reorganization of microtubules into the mitotic spindle apparatus, and it has been

postulated that extra centrosomes result in the unequal distribution of chromosomes [9]. A

pathogenic role has been identified for centrosome associated Aurora and Polo-like kinases.

STK15/BTAK/ARK1/aurora 2, a centrosome-associated serine-threonine kinase has been

showed to be amplified and overexpressed in colon cancer cell lines, and this STK15

overexpression induced aneuploidy in NIH3T3 cells [88, 89]. Polo-like kinase 1(Plk1), a

serine/threonine kinase, was demonstrated to be overexpressed in 73% of primary CRCs , and

Page 31: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

14

this correlated with tumour invasion, lymph node involvement and Dukes stage [90]. A recent

study implicated centrosome amplification in tumourigenesis when the transplantation of extra

centrosomes in the larval brain cells of flies induced the formation of metastatic tumours [91].

Telomeres refer to the segments of DNA bound by specific proteins that cap the ends of

chromosomes. Thus, telomeres act as a buffer to prevent the loss of valuable genomic sequence

during replication[92], as well as to prevent chromosomes fusing at the ends [76]. A RNA

primer is required for the process of DNA replication. Thus, when replication proceeds from the

5′3′ direction, it leaves a stretch of unreplicated DNA at the 5′ end. This leads to a gradual loss

of telomeric repeats and the consequent shortening of telomeres after each round of replication,

which is referred to as the „end-replication problem‟ [76]. A specific enzyme, telomerase

maintains telomere length. Telomerase consists of two main components; the reverse

transcriptase component (hTERT), which is only expressed in cells where telomerase activity is

present; and the ribonucleoprotein moiety (hTERC/hTR), which is expressed ubiquitously in all

cells. In adults, telomerase activity has been observed only in immature germ cells, certain

stem/progenitor cells and in a subset of somatic cells such as human fibroblasts. Upon reaching

a critical short telomere length, DNA damage checkpoints trigger senescence pathways and

apoptosis. Cells that survive this checkpoint activate telomerase or the Alternative Lengthening

of Telomeres (ALT) pathway. It has been proposed that it is therefore important for cancer cells

to regain the ability to maintain telomeres, in order to avoid senescence and extensive

chromosome fusion during crisis [93, 94]. Additionally, telomere loss has been shown to inhibit

tumour progression and the development of macroscopically advanced tumours [95-98]. It has

been estimated that about 85-90% of human cancers have reactivated telomerase and are able to

maintain telomere length[76] and increased telomerase activity has been observed in colorectal

tumours[99-101]. Interestingly cancer cells that are deficient for telomerase activity are able to

Page 32: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

15

maintain telomere length via ALT, possibly making use of DNA repair pathways and

recombination to maintain telomere length[102]. Thus, it appears that maintaining telomere

length is critical for tumourigenesis and cellular immortalization [76]. However, conflicting with

this view is the observation that the telomeres of invasive human cancers are often shorter than

their normal counterparts [103]. Furthermore, when the protection of telomere ends is

compromised, chromosomes enter breakage-fusion-bridge cycles that can dramatically

reorganize the genome [9]. Studies in telomerase deficient mice (mTERC-/-

) demonstrated that

telomere shortening led to increased rates of spontaneous tumour formation, initiation of aberrant

crypt foci and microadenomas in the gastrointestinal tract that[98]. Furthermore, telomeres from

colon cancer cells were found to be 77%-90% shorted than those in adjacent normal tissue [100,

104-106].Taken together this indicates that the timing at which the telomeres shortening occurs

plays a crucial role in cancer development [107]. Telomere shortening promotes CIN, which in

turns drives early carcinogenesis. In contrast telomerase activation and telomere stability confer

immortality during the latter stages of tumourigenesis [9].

Genomic stability is maintained by checkpoints that regulate the maintenance of DNA fidelity in

response to exogenous and endogenous genotoxic stress. In response to these stressors these

checkpoints initiate a cascade of events that culminates in either cell-cycle arrest, which provides

the cell sufficient time to repair the damage; or apoptosis/cellular senescence, if the damage

cannot be repaired. These DNA damage checkpoints are traditionally considered to belong to

two classes, the first group consists of checkpoints that recognize and respond to DNA damage

from genotoxic agents and the second is composed of those that monitor the fidelity of replicated

DNA [64, 108]. A number of DNA repair proteins have been demonstrated to play a role in

human cancers, including TP53, ATM, ATR, BRCA1 and BRCA2 and hRad17 [109-112]. Of all

these checkpoint proteins, TP53 has been the only one directly implicated in human colon

Page 33: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

16

cancer. However, it appears that while specific TP53 mutations cause CIN, the remaining

mutations likely affect other processes such as apoptosis, which has been postulated to create a

permissive state that allows CIN to occur but does not directly cause CIN [113]. Furthermore, the

types of rearrangements observed in primary human cancers are similar to those seen when these

checkpoint proteins are inactivated in yeast[114]. This observation provides indirect evidence for

a role of the replication checkpoints in the genomic instability of colon cancer.

In a subset of colorectal cancer cell lines, inactivation of the base excision repair (BER) system

appears to predispose to point mutations that contribute to CRC formation by causing genomic

instability at the level of the base pair [64]. BER is mainly responsible for repairing damage

induced by endogenous metabolic processes such as methylation, deamination, reactive oxygen

species (ROS) and hydrolysis [115]. Multiple proteins contribute to BER pathway and enable it

to correct non-bulky damaged nucleotides, abasic sites as well as single-strand breaks. The

process is initiated by DNA glycosylases specific for various types of damage, which recognize

and cleave the N-glycosylic bond that connects the damaged base to the DNA backbone [116].

To date, 11 such DNA glycosylases have been identified in mammals [117]. Biallelic

inactivation of one of the BER gene, MYH, leads to an inherited form of colorectal cancer known

as MYH-associate polyposis (MAP) [118]. Tumours with biallelic MYH germline mutations

demonstrate CIN but no show no differences in the frequency of mutations in genes such as

TP53, SMAD4, or TGFBR2, which suggest that they have a unique molecular pathogenesis

[119].

1.1.4.2 Microsatellite instability pathway (MSI)

Microsatellite instability (MSI) defined as a change of any length due to either insertion or

deletion of repeating units, in a microsatellite within a tumour when compared to normal tissue

Page 34: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

17

[120]. Microsatellites lengthen or shorten due to the increased propensity of DNA polymerases

to slip in these regions. Defective mismatch repair activity is associated with the MSI phenotype.

Instability of microsatellite repeats is seen 15% of sporadic CRCs, primarily due to

hypermethylation of the promoter of the mismatch repair gene, MLH1 [121, 122]. In addition,

tumours of as many as 85% of patients with Lynch syndrome (an inherited CRC syndrome) also

show MSI as a consequence of mismatch repair gene mutations [123, 124]. A number of

clinicopathological features have been linked with MSI-H, such as mucinous differentiation,

signet ring cell morphology, Crohn‟s-like reaction, increased tumour-infiltrating lymphocytes,

poor differentiation and a tendency to be located in the proximal colon [125-127]. Moreover, in

CRC the MSI-H phenotype is associated with a favorable prognosis and a decreased likelihood

of metastases [128].

Microsatellites are repetitive DNA sequences that occur throughout the genome. The functional

impact of resulting mutations is determined by their genomic localization. While these

sequences occur mostly in non-coding regions, short repetitive regions can also occur in the

coding regions of tumour suppressor genes. Mutations that occur within coding regions have the

potential to cause frameshift and truncating mutations, while those that occur within regulatory

intronic regions may potentially alter transcript level and stability. Recently, a public database of

mononucleotide repeat tracts mutated in microsatellite unstable human tumours was described

(http://www.seltarbase.org/) [129]. It is expected that this will facilitate the identification of

relevant genes in carcinogenesis driven by MSI. This database lists the multiple genes that

display somatic mutations in MSI-H tumours including TGFβRII, BAX, IGFIIR, BLM, CHK1,

caspase-5, MSH3 and MSH6. The impact and frequency of mutation in several of these genes is

briefly discussed below.

Page 35: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

18

It has been estimated that approximately 85% of CRCs with MSI have mutated TGFβ receptor II

(TGFβRII) [5, 130]. Somatic mutations that inactivate TGFβRII are mainly frameshift mutations

within the10 base-pair poly (A) repeat in the coding region (codons 125–128) [5]. Additionally,

protein truncation occurs due to insertion mutations in the poly (GT)3 microsatellite region in the

TGFβRII gene [5, 130]. In the colon, the ligand of TGFβRII, TGFβ1 potently inhibits the growth

of normal epithelial cells. Thus, mutated TGFβRII allows tumour cells to escape these growth-

suppressive effects.

Insulin like growth factor II receptor (IGFIIR) plays a critical role in cell growth, survival and

differentiation; however, once mutated, these receptors play a role in tumourigenesis [131]. The

IGFIIR interacts with both IGF-II and latent TGFβ1 ligands and acts as a tumour growth

suppressor. The IGFIIR gene harbors several microsatellites within its coding region, one of

these is a poly (G)8 repeat that is often mutated in MSI-H tumours [132].

Disruption of the APC/β-catenin pathway is a critical step in the development of CRC and other

cancers. The transcription factor Tcf-4 is a Tcf-family protein, which complexes with β-catenin

and plays an important role in maintenance of normal epithelial growth and development of

colorectal tumours. Frameshift mutations in the poly(A)9 mononucleotide repeat in exon 17 of

the Tcf-4 gene were found in MSI cells [133, 134]. This produces a short and medium isoform

of the Tcf-4 protein and the properties of these isoforms are still not clear. One possibility is that

mutated Tcf-4 protein may acquire increased transcriptional activity by increased binding

affinity for β-catenin. These mutated Tcf-4 proteins also lose the ability to bind chromatin

remodeling complexes and in doing so may facilitate the structural organization of chromatin

[135]. It has been suggested that mutations in Tcf-4 may contribute only marginally to the

colorectal carcinogenesis or perhaps even be passenger mutations [136].

Page 36: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

19

In endometrial and colorectal cancers, mutations in PTEN gene occur in MMR deficient cells

[137]. The loss of PTEN causes elevation of intracellular phosphatidylinositol 3-phsophate

(PIP3) levels that stimulate serine/threonine kinase Akt kinase activity. The active Akt leads to

tumourigenesis due to loss of proliferative and apoptotic controls of cell growth. The coding

region of the PTEN gene contains several microsatellites, among which a repeat of poly(A)6 tract

in exons 7 and 8, which are most susceptible to frameshift mutations[138].

Approximately 50% of the tumours with MSI have mutations in the Bax gene. These mutations

produce frameshift mutations within a coding region of eight nucleotide stretch of guanine

residues (G8) [139]. Bax is a pro-apoptotic protein that forms a heterodimer with BCL2, and

functions as an apoptotic activator. Bax is also thought to play a role in the release of cytochrome

C from the mitochondrion. Thus, mutations in BAX contribute to tumourigenesis by decreasing

apoptosis. MSI tumours also show somatic mutations in the MMR genes, MSH2, MSH3, which

may further exacerbate the MSI phenotype [140].

However, the panel of genes mutated in diverse cancers appears to be distinct. For example a

recent study found that mutations in of TGFβRII, BAX, IGFIIR, caspase-5, MSH3 and MSH6 are

rare in urothelial bladder carcinoma and are not associated with the MSI-H phenotype [141].

Furthermore, the temporal sequence of these mutations appears to be an important feature with

TGFβRII and Bax mutations reported to be later events in the adenoma to carcinoma sequence

[142, 143].

The MSI status of tumours can be determined by carrying out PCR on DNA from fresh or

formalin-fixed paraffin-embedded tissue. Individuals can show heritable and stable polymorphic

variation within microsatellite regions. Thus, differences in number of repeats at microsatellite

loci in tumour (abnormal) tissue can be only demonstrated when compared with normal tissue of

Page 37: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

20

the same individual [120]. The National Cancer Institute has recommended a panel of five

microsatellite markers for use in CRC, these include two mononucleotide repeats (BAT25 and

BAT26) and three dinucleotide repeats (D5S346, D2S123 and D17S250)[120]. A tumour that

shows instability in at least 2 of the 5 tested microsatellites is termed MSI-high (MSI-H), 1 of

the 5 is MSI-low (MSI-L), and 0 of the 5 is considered microsatellite stable (MSS)[120]. If more

that 5 markers are used to identify this phenotype then MSI-H is defined as instability at ≥30-

40% and MSI-L as instability at < 30%-40% of the markers. This panel may not be ideal for the

evaluation of MSI in endometrial carcinoma as significant differences in patterns of

microsatellite abnormalities are found between endometrial and CRC in families with MLH1 or

MSH2 germline mutations [144].

The independent existence of a MSI-L phenotype that is distinct from MSS has been contested in

the literature. For example, one study demonstrated that the majority of CRCs show some

degree of microsatellite instability when a large enough number of markers are tested, indicating

that the differences between the MSI-L and MSS subgroups is merely quantitative rather than

qualitative [145]. However, an emerging but significant body of work indicates that MSI-L is a

distinct biological entity and is not simply a PCR artifact [146, 147]. For example one study

distinguished MSI-L CRCs from the MSI-H and MSS subgroups based on gene expression

profiles[148]. MSI-L tumours also show frequent instability of the trinucleotide repeat region in

Ras-induced senescence 1 (RIS1) [149]. MSI-L status has also shown to be an independent

adverse prognostic feature in stage III CRC in patients not treated with chemotherapy [150],

particularly when associated with RIS1 mutation [149]. Despite these advances, further studies

are needed to determine the biological significance of this phenotype.

Page 38: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

21

MSI status may also serve as a predictive marker of the efficacy of certain chemotherapeutic

agents. For example, 5- Fluorouracil (FU) is a widely-used chemotherapeutic drug that initiates

apoptosis by targeting thymidine synthetase and/or by the direct incorporation of its metabolites

into DNA and RNA. Despite its success at treating colon cancer several recent studies have

suggested that 5-FU may not confer a survival advantage to stage II and III CRC patients whose

tumours are MSI-H compared to those with MSS tumours [151]. While these findings have

sparked some debate, emerging evidence indicates that the MSI status of a tumour is in fact an

important predictor of 5-FU response[152].

Other than the above mentioned mechanisms that are implicated in MSI, a recent study

demonstrated that the overexpression of microRNA (miR)-155 downregulated the MMR

proteins, MSH2, MSH6, and MLH1, consequently induced MSI [153]. miRs are noncoding

RNAs that play a role in the posttranscriptional regulation of over 30% of the human genes

controlling critical biological processes. Thus, miR-155 likely acts to destabilize the expression

of these MMR proteins. Furthermore, this study found an inverse correlation between the

expression of miR-155 and the expression of MLH1 or MSH2 proteins in human CRC. Previous

studies have noted that miR-155 is overexpressed in CRC [154, 155], and that this appears to be

more frequent in MSI than MSS tumours [156].

1.1.4.3 CpG island methylator phenotype (CIMP)

In addition to the chromosomal and genetic aberrations discussed above, epigenetic

modifications that affect both DNA and the associated chromatin are capable of influencing gene

expression and the stability of the genome. DNA methylation or the covalent modification of the

C-5 position of cytosine residues occurs primarily at the short stretches of CG dinucleotides

known as CpG islands. Recent estimates suggest that there are at least 29,000 such regions

in the

Page 39: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

22

human genome, many of which surround the 5' ends of genes [157]. In eukaryotes methylation is

thought to play a role in regulating gene expression and in silencing repeat elements in the

genome [158]. Methylation also plays an important role in inactivating one copy of the X

chromosome, so that equal gene dosage is maintained in the somatic cells of males and females

[159]. In normal cells the pattern of expression is stably maintained following DNA replication

and cell division by a maintenance enzyme, DNA methyltransferase (DNMT1). The first

epigenetic mechanism implicated in carcinogenesis was DNA hypomethylation (decreased

methylation) [160]. Age related decreases in DNA methylation levels occur in a tissue specific

manner [161, 162], and it is likely that these changes contribute to the age-related increase in

carcinogenesis [163]. CpG islands commonly occur in gene promoter regions, thus

hypermethylation (increased methylation) of this region has been shown to silence gene

expression [164]. Thus, given its ability to inactivate tumour suppressor genes DNA

hypermethylation has been widely studied in cancer and several genes including, MLH1, MGMT

and HLTF have been shown to be silenced by hypermethylation in CRC [64]. Furthermore, a

number of genes such as HLTF, SLC5A8, MGMT, MINT1, and MINT31 show aberrant

hypermethylation in aberrant crypt foci indicating that hypermethylation is an early event in the

adenoma-carcinoma sequence [165, 166].

In 1999 it was postulated that the subset of CRCs that demonstrate widespread CpG island

hypermethylation arose via a distinct pathway termed CpG Island Methylator Phenotype

(CIMP)[167]. However, the concept of CIMP was initially debated as it was not clear whether

CIMP tumours are a unique molecular subgroup, or if it was composed of tumours that fall on

the upper end of the continuum of aberrant DNA methylation; in other words if the only

distinguishing characteristic was merely quantitative differences in methylation[64].

Page 40: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

23

The initial report of CIMP used methylated in tumours (MINT) as markers of these tumours

[167]. Since then there has also been considerable research to discover the most suitable panel of

markers and at present it is not clear which markers will most accurately identify CIMP tumours.

This is further complicated by the finding that CIMP tumours are not a homogenous entity, and

CIMP has been classified into the subcategories CIMP-high (also referred to as CIMP-1),

CIMP-low (CIMP-2) and CIMP-0 [147]. For example, the methylation markers CACNA1G,

CDKN2A, BRABP1, IGF2, MLH, NEUROG1, RUNX3, and SOCS1 are specific for CIMP-high

but are not optimal for the identification of CIMP-low tumours[168]. In terms of biological

differences, CIMP-low was shown to be associated with KRAS mutation and male sex, CIMP-

high was associated with BRAF mutation and female sex, and CIMP-0 is associated with wild-

type KRAS and BRAF[168]. Furthermore in a study using a large number of methylation

markers, CIMP-high tumours formed a distinct group by unsupervised cluster analysis

suggesting that CIMP is a distinct subtype of CRC [169].

The mechanism by which CIMP arises is currently unknown but is the subject of much research.

However, it has been proposed that the synergy between KRAS and BRAF mutations in

combination with specific patterns of gene methylation is necessary to facilitate the early events

of tumourigenesis [147]. Additionally, it was recently proposed that all CRCs be classified based

on MSI and CIMP status. However, further studies will need to be undertaken prior to

implementing this classification [63, 147].

Page 41: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

24

A.

B.

C.

Figure 1.1 The simplified progression of CRC via the CIN, MSI and CIMP pathways.

Depicted by panel A, B and C respectively. Several key genes that are implicated in each

pathway are highlighted. Reprinted from Gastroenterology, Grady, WM and Carethers, JM.

Genomic and Epigenetic Instability in CRC pathogenesis, copyright 2008, with permission from

Elsevier [64].

Page 42: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

25

1.1.4.4 Cancer stem cell model and CRC

A cancer stem cell is a cell within the tumour that possesses the capacity to self-renew, and in

doing so gives rise to the heterogeneous lineages that comprise the tumour. Cancer cells may

arise therefore from tissue stem cells that have acquired mutations that render them cancerous, or

it may be a more differentiated (progenitor) cell that may have „re-acquired‟ stem cell like

properties due to mutations [170]. Cancer stem cells or cancer initiating cells have been

identified to date in acute myelogeneous leukemia[171], breast tumours [172], brain tumours

[173, 174] and most recently in a subset of colon tumours [175]. The colon cancer initiating

cells were able to induce human colon tumours that were phenotypically similar to the original

patient tumours after transplantation into non-obese diabetic (NOD)/severe-combined

immunodeficient (SCID) mice [175]. This cell population also self-renewed, which enabled

colon cancer to be re-established in secondary and tertiary recipient mice. A recent report

identified Lgr5+ cells, which are located at crypt bottoms as the putative stem cells of the small

intestine and colon [176]. Furthermore, deletion of Apc in these Lgr5+ cells led to the growth of

microadenomas that progressed to form adenomas in 3-5 weeks [177]. However, when Apc was

deleted in the short lived transit amplifying cells, adenomas were rarely seen even 30 weeks

later, which led the authors to conclude that the stem-cell specific loss of Apc results in a

progressively growing neoplasia [177]. Furthermore, a recent study found that high Wnt activity

functionally designates the colon cancer stem cell population[178]. Thus, based on this evidence

the adenoma to carcinoma model has been refined to suggest that the first mutation in APC

occurs in the gastrointestinal stem cell, which generates an APC -/+

mutant stem cell. These cells

will then progressively colonize the entire stem cell niche, while their progenitors take over the

crypts, an event that has been termed monoclonal conversion [9]. Once the second APC allele is

lost, niche succession occurs again and the niche fills with APC -/-

cells. The subsequent

Page 43: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

26

acquisition of downstream mutations in KRAS and TP53 will then occur within this cancer stem

cell population.

1.1.5 Behavioural and lifestyle risk factors

Multiple studies have attempted to evaluate the relationship between CRC risk and the intake of

specific dietary compounds. However, several of these studies have produced conflicting data

and the precise involvement of dietary factors in decreasing or increasing the risk of CRC is

currently unclear. Several epidemiologic studies have shown an association between a diet low

in fruits and vegetables and increased risk of CRC [179, 180]. Furthermore, the relative risk of

CRC was found to be approximately 0.5 in groups with the highest vegetable and fruit intake

compared to those with the lowest [179]. Also a recent systematic review of the literature

indicates that the majority of available evidence supports the association of red meat

consumption with an increased risk of CRC [181, 182]. PhIP (2-amino-1-methyl-6-

phenylimidazo[4,5-b]pyridine) is a bulky adduct forming agent that is commonly found in well-

done meat and poultry and has been shown to cause a variety of cancers in experimental animals

[183-186]. Another powerful rodent carcinogen is MNNG (N-methyl-N'-nitro-N-

nitrosoguanidine), is an alkylating agent that is able to preferentially methylate the O6

position of

deoxyguanosine residues in DNA [187]. Gastrointestinal cells are continually exposed to both

PhIP and MNNG at varying concentrations. In a study undertaken to determine if carcinogen

exposure can influence the type of instability seen in cells, it was found that cells resistant to

PhIP developed a chromosomal instability or CIN phenotype. Conversely, cells resistant to

MNNG exhibited MSI and associated mismatch repair defects [188]. This suggests that exposure

to certain dietary carcinogens, may in fact select for cancer cells with distinct types of genetic

instability, however further work is needed to confirm these findings [188].

Page 44: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

27

An association between heavy alcohol consumption and increased risk of CRC has been

reported in several studies [189, 190]. It has been estimated that the CRC risk was increased

slightly (relative risk of 1.41) in individuals who consumed over 45g of alcohol per day, and this

risk was slightly lower for those who consumed between 30 to 45g per day [191]. Interestingly,

alcohol consumption was found to be associated with an increased risk of cancers of the distal

colon and rectum but not with cancers of the proximal colon[192]. The specific mechanism

linking ethanol and carcinogenesis is not clear. However, it is believed that the major toxic

byproduct of ethanol metabolism, acetaldehyde is capable of inducing point mutations,

chromosomal aberrations and alterations in protein structure [193].

Furthermore, multiple studies have examined the contribution of lifestyle factors such as

smoking and physical activity to CRC incidence. Decreased regular physical activity is

associated with increased risk of CRC, an effect that has been proposed to be linked to high

energy intake, obesity, and the metabolic syndrome [194]. However, the precise mechanism by

which physical activity exerts its protective effect is currently unclear [182]. In a recent meta-

analysis smoking was associated with an increased risk of CRC incidence and mortality[195].

This study also found that this risk increased with increasing dose (increased number of pack

years and cigarettes per day), but this association was only statistically significant after 30 years

of smoking. Interestingly, with respect to both incidence and mortality, the association was

stronger for cancer of the rectum than of the colon; suggesting that unique pathological processes

at play at distinct anatomic sites.

Patients with chronic inflammatory bowel diseases involving the colon such as ulcerative colitis

or Crohn‟s disease are at significantly increased risk for CRC [196, 197], and special

colonoscopic surveillance measures are recommended for such individuals [198]. Several

Page 45: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

28

factors may influence the risk of CRC in patients with inflammatory bowel disease, including

earlier onset of disease (prior to 15 years) and the severity of inflammation [182, 199].

1.1.6 Hereditary syndromes, pathology and presentation

The most common form of inherited CRC is Lynch syndrome (MIM# 120435), also referred to

as hereditary nonpolyposis colorectal cancer (HNPCC). Due to its relevance to this thesis, this

syndrome will be discussed in more detail in section 1.2.

Familial adenomatous polyposis (MIM# 175100) is an autosomal dominant disorder that is

caused by germline mutations in the APC gene [200]. In this syndrome multiple adenomas occur

at an early age, often during preteen years and proliferate throughout the colon with malignant

degeneration by the age of 40 - 50 years. Adenomas in the duodenum, which carry the risk of

progressing to periampullary carcinoma are also seen in FAP [201]. Furthermore, some

individuals especially those of Korean or Japanese descent also have an increase likelihood of

adenomas and cancers of the stomach [202]. Most mutations detected in this gene are truncating

or frameshift mutations and a smaller proportion are missense in nature. The APC tumour

suppressor gene acts in a nonclassical manner in that the localization of the germline mutation

determines the position as well as the type of second hit likely to occur [203, 204]. Moreover, an

association was also found between the location of the mutation, the number of colonic polyps

and the severity of the disease. For instance, premature stop codons that occur between codon

1250 and 1464 cause a profusion (about 5000) of polyps, while mutations that are upstream or

downstream of this region are associated with a later onset of disease and lead to a lesser number

of polyps (about 1000)[205]. The penetrance of this syndrome varies, with penetrance

approaching 100% in classic FAP caused by germline mutations in the APC gene. However,

penetrance is only about 10-20% for the I1307K APC polymorphism in Ashkenazi Jews. An

Page 46: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

29

attenuated form of this disease occurs when mutations occur at the extreme 5‟ end, the extreme

3‟ end or in exon 9 of APC [206, 207]. These individuals had comparatively fewer polyps (<50-

100), and the polyps tended to occur one to two decades later in life than classical FAP. Gardner

syndrome represents one tail of the FAP phenotype. Gardner syndrome represents the association

of colonic adenomatous polyposis of classic FAP with osteomas and soft tissue tumours

(epidermoid cysts, fibromas, desmoid tumours). APC mutations associated with Gardner

syndrome tend to occur between codons 1,403 and 1,587 [208].

Germline mutations in the BER gene, MutY human homologue (MYH) leads to an inherited form

of colorectal cancer known as MYH-associated polyposis (MAP; MIM# 608456)[118]. It has

been approximated that MYH is the cause of adenomatous polyposis in up to 5%–10% of

individuals who have FAP. The identification of an excessive number of somatic G:C A:T

mutations in neoplasms of people with adenomatous polyposis with no detectable germline

mutations in APC made way for this discovery [118, 209, 210]. This is due to the ability of

reactive oxygen species to modify the C8 position of Guanine to form 7, 8-dihydro-8-oxoguanine

(8-oxoG), a highly mutagenic substrate that is able base pair with adenine and cause G:CT:A

transversions [211]. Interestingly, two common missense mutations Y179C and G396D

(formerly described as Y165C and G382D) account for the majority (~80%) of mutations

discovered in MYH [118, 210, 212, 213]. Furthermore, compared to noncarriers monoallelic and

biallelic carriers of these germline mutations have an increased risk of CRC and an increased

likelihood of having first-and second-degree family members with CRC [212]. Despite the

identification of germline mutations in MYH, somatic MYH mutations are not common in

sporadic colorectal cancer (203, 204).

Page 47: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

30

There are also several rare hamartomatous polyposis syndromes that predispose to CRC.

Germline mutations in the PTEN gene are responsible for phenotypically diverse conditions

which have are collectively referred to as the PTEN hamartoma tumour syndrome (PHTS). One

disease that belongs to this subset is Cowden syndrome (CS, MIM#158350), a rare autosomal

condition that is difficult to diagnose [214] The most important and universal indicator of CS is

mucocutaneous features. Over 60% of those affected with CS are thought to develop

hamartomatous polyps in the GI tract [215] and while colon cancer has been reported, its

involvements in CS has not been confirmed [216]. Although originally thought to be distinct

conditions, Bannayan-Riley-Ruvalcaba syndrome (BRRS, MIM#153480) is now believed to be a

single entity. In the absence of definitive clinical criteria, it has been suggested that individuals

with combinations of macrocephaly, lipomatosis, hemangiomas, GI polyposis, and pigmented

macules of the penis be considered clinically affected[217]. Furthermore, GI, breast, thyroid and

endometrial cancers are thought to be features of this syndrome[218].

Peutz-Jeghers (PJS, MIM#175200) is an autosomal dominant syndrome associated with

mucocutaneous hyperpigmentation and benign intestinal polyps known as hamartomas. The

characteristic pigmented macules are benign and are not thought to have malignant potential.

Approximately 50% will have experienced gastrointestinal symptoms by age 20 [219]. Unlike

other hamartomatous syndromes, PJS related polyps occur most frequently in the small intestine.

These individuals are at a high risk of developing malignancies especially of the breast and colon

[220, 221]. A majority (60-70%) of Peutz-Jeghers patients show germline mutations in the

serine/threonine protein kinase STK11 (also known as LKB1) [222].

Juvenile polyposis syndrome (JPS, MIM#174900) often presents before the age of 20 and is

characterized by the development of multiple juvenile polyps in the gastrointestinal tract.

Page 48: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

31

Approximately 25%-50% of JPS cases are believed to be de caused by de novo mutations[216].

Deletions in SMAD4 and BMPR1A, both components of the TGFβ pathway account for a

significant proportion of JPS-causing mutations [223]. Approximately 20% of individuals with

JPS are thought to have mutations in SMAD4, and a similar proportion of JPS is thought to arise

due BMPR1A mutations [224]. Individuals with JPS are at risk of developing cancers in the

stomach, small intestine and colon.

1.2 Lynch Syndrome

1.2.1 Overview

Germline mutations in mismatch repair genes are associated with Lynch syndrome or hereditary

nonpolyposis colorectal cancer (HNPCC). Lynch syndrome, an autosomal dominant disorder that

accounts for about 2% of all CRC cases, is one of the most common cancer predisposition

syndromes. Instability of microsatellite repeats is seen in tumours of as many as 85% of patients

with HNPCC, making it a hallmark feature of this syndrome [123, 124]. Five cardinal features

have been suggested to help identify families affected by Lynch syndrome [225]; 1) an earlier

age at cancer onset compared to the general population (estimated to be ~ 45 years[226]

compared to ~63 years), 2) the pattern of primary cancers segregating in the family, especially

CRC and endometrial cancer, 3) survival rates that differ from the norm, 4) the identification of

distinguishing pathological features associated with the tumours and 5) the identification a

germline mutation in the affected family members.

Page 49: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

32

1.2.2 Pathology, presentation and clinical criteria

In Lynch syndrome multiple generations are affected with colorectal cancer at an earlier age with

a predominance of right-sided CRC (approximately 70% are proximal to the splenic flexure).

There is also an excess of synchronous CRC (defined as multiple CRCs at or within 6 months

after surgical resection for CRC) and metachronous CRC (CRCs occurring more than 6 months

after surgery) [226]. Additionally, there are several distinguishing pathological features that set

apart Lynch syndrome tumours [127, 227-229]. Compared to sporadic CRC, Lynch syndrome

tumours are more often poorly differentiated, with an excess of mucoid or signet ring features.

They are also characterized by a Crohn‟s-like reaction (lymphoid nodules, including germinal

centers, located at the periphery of the infiltrating tumour) as well as the presence of infiltrating

lymphocytes within the tumours. Furthermore, Lynch tumours undergo accelerated

carcinogenesis. For example, a small adenoma may emerge as a carcinoma is 2-3 years

compared to the 8-10 years this process takes in sporadic cancers [227].

In the clinical setting, diagnosis of Lynch syndrome is complicated by the lack of precise

characteristic features. Clinical guidelines based on family history and age of onset of CRC,

known as the Amsterdam criteria-1, were initially developed and used to identify putative Lynch

families [230] (Table 1.4). These criteria were later modified to include extra-colonic cancers

that were frequently seen in these Lynch kindreds (Amsterdam criteria II) [231]. The realization

that Amsterdam criteria did not capture all individuals with Lynch syndrome led to the

development of Bethesda criteria. These criteria help determine if individuals from families that

may not meet Amsterdam criteria should undergo genetic testing for Lynch syndrome [232,

233]. Although widely used to aid in diagnosis, these criteria are limited in their ability to

accurately identify patients with Lynch syndrome. Thus, the diagnosis relies primarily on the

Page 50: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

33

detection of germline defects in the MMR genes of these patients [234]. It has been estimated

that 40-80% of Lynch syndrome families that meet Amsterdam I criteria and 5-50% of families

meet Amsterdam II criteria carry mutations in the MMR genes [235, 236]. Hitherto undiscovered

genes, shared environmental influences or chance can account for families that do not show

MMR mutations, as well as families that show clustering of Lynch syndrome associated cancers

but do not meet these formal criteria [237]. Furthermore, the subset of families that meet

Amsterdam criteria-1 but do not show MMR deficiency have been recently classified as a

distinct syndrome known as familial colorectal cancer type X [238]. The risk of CRC is

significantly decreased for these individuals compared to those in families with Lynch syndrome

Page 51: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

34

Table 1.4 Summary of clinical criteria used to identify Lynch syndrome

Amsterdam Criteria I [230]

-To help identify Lynch

syndrome families

1. At least 3 relatives should have histologically verified

CRC

2. One should be a first-degree relative of the other 2

relatives

3. At least 2 successive generations should be affected

4. At least 1 relative should be diagnosed before age 50

years

5. Familial adenomatous polyposis should be excluded

Amsterdam Criteria II

[231]

- To help identify Lynch

syndrome families that may

have been excluded due to

the stringency of the above

criteria

1. At least 3 relatives should have cancers associated with

the Lynch syndrome

2. One should be a first-degree relative of the other 2

relatives

3. At least 2 successive generations should be affected

4. At least 1 relative should be diagnosed before age 50

years

5. Familial adenomatous polyposis should be excluded

6. Tumours should be verified whenever possible

Revised Bethesda Criteria

[239]

- To determine if tumours

should be tested for the MSI

Phenotype

1. CRC diagnosed in a patient who is younger than 50 years

2. Presence of CRCs that are synchronous or metachronous

or other tumours associated with Lynch syndrome

(regardless of age)

3. CRC with MSI-H or histology diagnosed in a patient who

is younger than 60 year

4. CRC or tumour associated with Lynch syndrome

diagnosed before age 50 years in at least 1 first-degree

relative

5. CRC or tumour associated with Lynch syndrome

diagnosed at any age in 2 first- or second-degree relatives

Page 52: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

35

1.2.3 Tumour spectrum

In Lynch Syndrome families an individual‟s lifetime risk of developing colorectal cancer (CRC)

increases to about 80% [233]. In addition to cancer in the colon, Lynch syndrome is

characterized by extra-colonic sites including the endometrium, stomach, urinary tract, ovaries,

small bowel, hepatobilliary tract, pancreas and brain, with endometrial and ovarian cancer being

the second and third most common cancers in these families [240, 241]. Individuals with Lynch

syndrome may also present with sebaceous adenomas/carcinomas and keratoacanthomas, a

finding that is consonant with the Muir-Torre variant (see section 1.3.5)[226].

1.2.3.1 Endometrial cancer

The cumulative lifetime risk of developing endometrial cancer has been estimated to be about

40-60% for women in Lynch syndrome families, which equals or exceeds their risk of CRC

[242]. This risk is higher in those with germline mutations in the MSH6 gene, and was estimated

to be about 70% by the age of 70 [243]. The average age for the development of endometrial

cancer in Lynch syndrome is around age 50 (mean

age of diagnosis is over 60 years in the

general population) [244]. Over 75% of women with Lynch syndrome who develop endometrial

cancer present with stage 1 disease, similar to sporadic disease [244].

MMR deficiency has also been shown to give rise to sporadic endometrial cancer and about 28%

of all endometrial tumours display the MSI phenotype primarily due to MLH1 promoter

hypermethylation [245, 246]. Endometrial cancer is the fourth most common cancer in women

in Canada, with an estimated incidence of 4500 cases in 2010 [20]. For those at average risk the

lifetime probability of developing endometrial cancer is about 2.4% and the estimated five-year

relative survival ratio from 2002-2004 was about 85% [21]. Although MSI-high CRCs are

Page 53: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

36

generally associated with a more favourable prognosis, the clinicopathologic impact of MSI in

endometrial cancers is not clear. A recent study found that the MSI-H phenotype is associated

with a worse prognosis in women with early stage( I/II) disease [247]. However, more studies

with larger patient cohorts are required to confirm this finding.

1.2.4 Predictive genetic testing

Advances in the understanding of the genetic basis of Lynch syndrome has allowed the clinical

community to initiate and utilize predictive genetic testing programs to accurately assess disease

risk for patients presenting with a family history of colorectal cancer. These programs help

identify individuals with an inherited MMR mutation who are at an increased risk of developing

colorectal and other Lynch syndrome associated cancers. By predicting risk prior to the

progression of disease, these programs play an integral role in the effort to improve surveillance,

treatment and prevention of cancer. One of the most important steps that will help with the

diagnosis of a hereditary cancer syndrome, including Lynch syndrome is the compilation of a

thorough family history of cancer [225, 248]. If the presence of such a syndrome is identified

then molecular genetic testing will provide verification of the diagnosis [249]. The diagnosis of

Lynch syndrome relies primarily on the identification of germline defects in the MMR genes

[234]. The Ontario Predictive Genetic Testing Program screens individuals presenting with either

a family history of colorectal cancer, and/or those who develop tumours at a young age of onset

(<50). Genetic screening is initiated by performing MSI testing and immunohistochemistry

(IHC) for the MMR proteins on tumour specimens when available. This is followed by germline

mutation testing on lymphocyte DNA. Initially, multiplex ligation-dependent probe amplification

(MLPA) is performed to detect large genomic deletions and duplications in the MMR genes. If

such changes are not detected, germline DNA is screened by sequencing the MLH1 and MSH2

Page 54: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

37

genes, guided by MSI/IHC results of patient tumours. Once diagnosis of Lynch syndrome is

established the proband‟s high-risk relatives will be notified and genetic counseling and

appropriate genetic testing will be performed on consenting individuals. Thereafter, surveillance

measures are put in place, in attempt to reduce morbidity and mortality of the associated

cancers[226]. For CRC the screening measures recommended are full colonoscopy every 1-3

years initiated between the ages of 20 and 25 for those with strong clinical evidence/ documented

germline mutations [225, 250]. Furthermore, it is recommended that females in Lynch syndrome

families be screened for endometrial and ovarian cancer, given the observance of accelerated

carcinogenesis in such cancers. For endometrial cancer, annual transvaginal ultrasonography and

endometrial aspiration for pathological assessment should begin at age 30. Screening measures

for ovarian cancer should include transvaginal ovarian ultrasonography and CA 125 screening

beginning at age 30. However, the screening methods available for the ovary are low in

sensitivity and specificity [225]. Screening for CRC in Lynch syndrome families has been show

to halve the risk of CRC and reduce the overall mortality associated with this disease by about

65% in these families [251].

1.3 Mismatch Repair

Mismatch repair (MMR) plays a central role in maintaining the integrity of genetic material and

the basic features of this system have been highly conserved throughout evolution. The best

understood function of the MMR system is the post-replicative repair of DNA polymerase errors.

Page 55: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

38

1.3.1 Overview of the mismatch repair system in prokaryotes

The mismatch repair proteins were first identified in prokaryotic organisms and the Escherichia

coli (E.coli) MMR system has been completely reconstituted in vitro. MutS is an ATPase that

recognizes and binds (as a homodimer) to base mismatches and insertion/deletion loops [252].

The C-terminal end of MutS contains a P-loop motif for nucleoside triphosphate binding, and

ATP binding/hydrolysis promotes disassociation of the MutS complex. In E.coli newly

synthesized DNA is transiently unmethylated and this provides the signal that facilitates strand

discrimination. MutH, a methylation sensitive endonuclease cleaves the unmethylated strand of a

hemimethylated GATC dam methylation site, thereby targeting the nicked strand for

exonucleolytic removal and repair. MutH can nick DNA on either side of the mismatch and

depending on the side on which cleavage occurs exonucleases with either 5‟3‟ polarity (RecJ

or ExoVII) or 3‟5‟ polarity (ExoI or ExoX) polarity excise the nicked strand [253, 254]. These

nucleases are single strand specific, therefore UvrD helicase acts to unwind the duplex molecule

in a directional manner beginning at the nick towards the mismatch [255]. The strand is then

resynthesised by DNA Polymerase III, following which the nick is sealed by DNA ligase.

MutL couples mismatch recognition by MutS to the downstream processing steps; however, its

precise role in this process has been difficult to decipher. The N-terminus of MutL is composed

of ATP hydrolysis/binding domains and is highly conserved [256]. The C-terminal region is

thought to contain the domain important for MutL dimerization. MutL has been shown to

directly interact with MutS by affinity purification[257]. Furthermore, the MutL directly

interacts with MutH and the interaction of MutL alone has been shown to be able to stimulate the

endonuclease activity of MutH[258]. MutL also is able to directly interact with and activate the

activity of the UvrD helicase[259]. Thus, based on these studies it has been proposed that MutL

Page 56: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

39

couples the mismatch recognition/binding activity of MutS, with the helicase activity of UvrD

and the mismatch specific strand cleavage activity of MutH.

1.3.2 Overview of the mismatch repair system in eukaryotes

All eukaryotes studied to date possess multiple MutS homologs (MSH proteins) and MutL

homologs (MLH proteins). Unlike their prokaryotic counterparts these proteins form

heterodimers with other MSH or MLH proteins. The MMR system in yeast is very well

characterized and the insights gained from these studies have been helpful in elucidating the

mammalian MMR system. All yeast MMR proteins, with the exception of the mitochondrial

protein MSH1 have been identified in mammals. Furthermore no homologs of MutH or UvrD

have been identified in eukaryotes. The central MutL homolog, MLH1 forms heterodimers with

the three remaining MutL homologs PMS1 (post-meiotic segregation protein 1), PMS2 and

MLH3 (Figure 1.2). The MLH1-PMS2 complex, referred to as MutLα appears to be the major

player in MMR [260]. The roles of the two other MutL complexes, MLH1-PMS1 (MutLβ) and

MLH1-MLH3 (MutLγ) in post-replicative error repair remains largely inconclusive, although it

believed that each could act as a substitute for MutLα if the need arose [261, 262]. MutLγ has

been proposed to be primarily involved in meiotic recombination [263].

The mammalian MSH2 protein forms two heterodimeric complexes. The first complex, MSH2-

MSH3 (MutSβ) preferentially recognizes insertion/ deletion loops; and the second complex,

MSH2-MSH6 (MutSα) recognizes both base-base mispairs and small insertion/deletion loops

[264-266]. Furthermore, the stability of both MSH3 and MSH6 depends on the presence of

MSH2 [267]. Like their yeast counterparts, the mammalian MSH4 and MSH5 proteins appear to

be meiosis specific [268, 269].

Page 57: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

40

Figure 1. 2 Heterodimers formed by the MutL homologs

Mismatch repair

Mismatch repair? - Mismatch repair?

- Meiotic recombination

Recognizes

mismatches

& IDLs

Recognizes

larger IDLs

Page 58: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

41

1.3.2.1 Mouse models of mismatch repair

Several mouse models of mismatch repair have been developed and this has helped us gain a

deeper appreciation of their roles in mammals. Mlh1-null (Mlh1−/−

) mice display a wide spectrum

of tumours, including lymphomas and intestinal tumours, increased MSI, and decreased survival

[270]. The Pms2−/−

mice also showed tumour susceptibility, albeit a less severe phenotype than

observed in the Mlh1−/−

mice, but did not develop gastrointestinal tumours [271]. Therefore it

was proposed that there must be proteins that serve as substitutes for PMS2. Mlh3−/−

mice

developed adenomas and carcinomas in the intestinal tract and showed weak instability in

mononucleotide microsatellites [272]. The slightly higher mutation rate and tumour burden in

Mlh1−/−

compared to Pms2−/−

mice is evidence of the minor role of MutLβ in mammalian MMR

[271, 273]. Mlh3−/−

and Pms2−/−

double-null mice showed increased tumour susceptibility and

reduced survival, similar to the results observed in Mlh1−/−

mice[272].

The phenotypes observed in Msh2, Msh3 and Msh6-null mice can be explained by the partial

redundancy between MutSα and MutSβ heterodimers [274]. Of these the Msh2−/−

mice displayed

the most severe phenotype; they developed multiple tumours in the colorectal, gastric, and

endometrial regions, along with lymphoblastic lymphomas, and survived for only 2 to 5 months

[275]. This is in line with MSH2s involvement in both MutS heterodimers. Msh6−/−

mice also

developed a spectrum of tumours, mainly of the gastrointestinal tract, and lymphomas but did not

show any signs of MSI [276]. The Msh3−/−

mice did not show a predisposition to CRC and

developed tumours at a late age [277]. However, at 2 years the tumour incidence of Msh3−/−

mice

was comparable to Msh3+/+

mice. The milder phenotype associated with the Msh3−/−

is likely due

to the ability of MutSα to recognize and bind to most types of mismatches and a subset of

insertion deletion loops. Mice with double homozygous deletions (Msh3−/−

/Msh6−/−

) had an

Page 59: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

42

increased predisposition to intestinal tumours, decreased survival, and loss of MMR activity

[278]. Deletions of the other MutS homologs Msh4 and Msh5 failed to show any predisposition

to cancers or decreased survival.

1.3.3 The post-replicative error repair function

The post-replicative error repair function is capable of eliminating base-base mismatches and

insertion/deletion loops that arise during DNA synthesis and have escaped the proofreading

activity of the DNA polymerases. The MMR replication error repair improves the fidelity of

DNA biosynthesis 100-1000 fold and reduces the error rate to one error per 1010

bases[279]. In

addition to the MutS and MutL homologs this function requires the involvement of several other

factors, most of which are involved in DNA replication. The first such factor, Exo1 is the only

enzyme with 5‟3‟ directionality that been implicated in MMR [274]. This was an intriguing

finding as the strand break can be either 5‟ or 3‟ of the mispair, thus MMR was thought to

require exonucleases with both 3‟5‟ and 5‟3‟ polarity. However, this disparity was resolved

by the finding that the PMS2 subunit of MutLα possesses a latent endonuclease activity that can

introduce single-strand DNA breaks in the discontinuous strand, thereby generating an entry

point for Exo1[280]. Exo1-/-

mice are prone to lymphomas but do not show gastrointestinal

tumours [281]. Furthermore, the embryonic stem cells of these mice also show a weak mutator

phenotype, confirming the role of Exo1 in MMR.

Another key protein implicated in MMR is proliferating cell nuclear antigen (PCNA). PCNA is

loaded on to primer-template junctions by the replication factor C (RFC). Once loaded the

PCNA sliding clamp functions as the processivity factor of the DNA polymerases. PCNA was

also shown to directly interact with MSH3, MSH6, MLH1 and EXO1 [282, 283]. The interaction

with MutSα and MutSβ via MSH6 and MSH3 indicated PCNA‟s involvements in steps

Page 60: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

43

preceding DNA synthesis and the presence of the MutS complexes in the DNA replication

complex. Furthermore, binding of MutSα to mismatches leads to its dissociation from PCNA

[284]. Therefore, it is likely that MutSα is targeted to the replication fork by binding to PCNA,

then transferred from PCNA to the DNA upon encountering a mismatch [284].

The initial steps in mammalian MMR are very similar to the repair process in prokaryotes. MMR

begins with the recognition of the mismatch or the insertion-deletion loop by MutSα or MutSβ

(Figure 1.3). This is bound by MutLα which functions to recruit other proteins involved in this

process. When this complex encounters PCNA, the binding of PCNA/RFC leads to the activation

of a MutSα /PCNA/RFC/ATP-dependent endonuclease activity of MutLα [280]. This generates

nicks in the discontinuous strand which function as entry points for EXO1. The rate of DNA

resection by the MutSα -EXO1 complex is controlled by the replication protein A (RPA), which

binds and protects the single-stranded DNA. Multiple cycles of mismatch-dependent reloading of

MutSα and EXO1 and 5‟3‟ degradation is thought to occur until the segment containing the

mismatch is degraded [285]. It is presently believed that MutLα participates in the termination

of this excision step. Following nucleolytic excision of the mismatch, repair synthesis is

accomplished by DNA Polymerase together with PCNA[286]. The final step of the mismatch

repair process involves sealing of the nick by DNA ligase I. The complete MMR process was

reconstituted from purified recombinant constituents by supplementing the MutSα, MutLα, RPA,

EXO1, PCNA, RFC (HMGB1) system with DNA polymerase and DNA ligase I [285, 287]. In

both these cases the reconstituted system catalyzed the repair of a heteroduplex substrate that

contained a G/T mismatch and a 5' strand break. In addition, in one of the systems replacing

MutSα with MutSβ allowed the system to efficiently repair an IDL of three nucleotides [285].

Page 61: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

44

Figure 1.3 Simplified overview of the eukaryotic MMR system.

Key proteins implicated in this process are highlighted. Reprinted by permission from Macmillan

Publishers Ltd: Nat Rev Mol Cell Biol, Jiricny J. The multifaceted mismatch-repair system,

copyright 2006[274].

Page 62: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

45

Nevertheless there still some uncertainties regarding the model of eukaryotic MMR. For

instance, while the transient hemimethylation of the nascent DNA strand facilitates strand

discrimination in prokaryotes, it is not clear how eukaryotes ensure that DNA repair is directed

towards the newly synthesized DNA strand. However, it was postulated that replication-

associated strand discontinuities may serve this purpose in vivo [288]. The molecular mechanism

of mismatch recognition is also currently unclear. Based on the conservation of DNA binding

motifs it appears that only MSH6 is involved in the initial mismatch recognition[289]. Thus, how

MutSβ recognizes IDLs is currently unknown. Furthermore, findings indicate that mismatch

binding in the presence of ATP and magnesium brings about a rapid conformational change in

MutSα, which allows it to release the mismatch and move along the DNA in the form of a sliding

clamp [274]. Three models of how mismatch recognition is coupled with downstream repair

processes have been have been proposed, the molecular-switch model, active-translocation

model and the DNA bending/verification model. The molecular-switch model proposes that

multiple MutSα (possibly in complex with MutLα) sliding clamps diffuse bidirectionally from

the mismatch [290]. When a clamp encounters a strand break tagged with other MMR factors

such as PCNA it will trigger the repair process. The active-translocation model postulated that

MutSα (possibly in complex with MutLα) will translocate along the DNA in a controlled manner

that is dependent on ATP hydrolysis once it is released from the mismatch [291]. The DNA

bending/verification model proposes that MutS complexes remain in the vicinity of the

mismatch, and that communication between the mismatch and the strand-discrimination signal

involves DNA bending rather than protein movement along the DNA[292]. This last model is

Page 63: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

46

based on studies in E. coli and has thus far not been demonstrated in a mammalian system.

Thus, current evidence favours the first two models [274].

1.3.4 The role in signaling DNA damage

MMR proteins have also been implicated in signaling DNA damage and apoptosis [293]. MMR

proteins can act on DNA damage caused by oxidation, heterocyclic amines, cisplatin, UV

radiation, ionizing radiation, methylating agents and alkylating agents i.e. N-methyl-N '-nitro-N-

nitrosoguanidine (MNNG), N-methyl-N-nitrosourea (MNU) and its analogues used in

chemotherapy. Recognition and perhaps processing of these lesions by MMR proteins leads to

activation of damage signaling pathways, which can in turn result in cell-cycle arrest, and even

apoptosis. Although the role of the individual MMR proteins in post-replicative DNA repair is

relatively well defined, their contributions to these other functions are not as well understood.

Based on previous experiments two models have emerged to explain the role of MMR proteins

in signaling DNA damage [294, 295]. In the first model, it is proposed that MMR proficient

cells‟ repeated futile attempts to remove mismatched bases alongside the damaged base leads to

DNA strand breaks. Cellular sensors of DNA damage recognize these breaks, and this

consequently leads to apoptosis. However, current experimental evidence has not been able to

directly substantiate this hypothesis. The second model, which postulates that MMR proteins

may be able to directly sense damage and trigger cell cycle arrest and apoptosis, without the need

for further processing was put forth based on later experimental evidence.

MMR-deficient cells are around 100 times more resistant to killing by alkylating agents and

around 2-4 fold more resistant to killing by cisplatin [295]. The model alkylating agent MNNG

and its analogue in clinical use, temozolomide, are able to at lower doses methylate the O6

Page 64: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

47

position of guanine, to form O6

methylguanine (O6

MeG). In the absence of functional MMR the

O6

MeG lesions persist and DNA damage accumulates but does not trigger cell death. This

causes cells to be tolerant to the cytotoxic effects of methylating/ alkylating agents [295].

Previous studies have illustrated that MMR proteins may be able to cause G2/M arrest [296] or

cell death[297, 298] in response to alkylating agents. This G2 checkpoint, and sensitivity to these

agents can be restored by the transfer of human chromosome 3 (normal location of MLH1) or

chromosome 2 (MHS2), into cancer cell lines that lack functional MLH1 or MSH2 respectively

[299, 300]. Treatment of cells with MNNG has been shown to phosphorylate p53 at Serine 15

and Serine 392 in a MMR protein dependent manner. These residues of p53 are preferred

substrates of the ATR and ATM kinases, which play critical roles in parallel DNA damage

pathways [301, 302]. Furthermore, exposure to alkylating/ methylating agents have been

reported to involve MutSα and MutLα dependent phosphorylation of CHK1 and CHK2, further

implicating their upstream kinases ATR and ATM in these processes [303-306]. More recent

evidence indicates that DNA damage results in the stabilization and accumulation of the MutL

proteins, MLH1, PMS2 and PMS1 in an ATM dependent manner [307]. This study was able to

establish that MLH1 is required to augment this ATM dependent phosphorylation of p53,

potentially indicating the role of MMR proteins as regulators of the amplitude of the p53

response. This is in contrast to other studies that suggest that while p53 is stabilized in response

to MNNG, MMR dependent apoptosis can occur independent of p53 expression [298]. In

addition to the above pathways, in MMR proficient glioma and colon cancer cells, temozolomide

was shown to activate the mitogen-induced p38α kinase and the G2 checkpoint, while no

activation was observed when the glioma cells were treated with siRNA against MLH1[308].

Although the exact mechanisms by which MMR proteins are linked to the signaling pathways is

unknown, collective evidence supports the notion that MMR protein likely play an important role

Page 65: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

48

in signaling cell cycle arrest and apoptosis (a simplified model of MMR dependent damage

signaling network is depicted in Figure 1.4).

Previous studies have suggested that the decreased levels of MMR proteins might promote

tumourigenesis. For instance Msh2+/-

mice are cancer prone and present with tumours that still

retain the wild-type copy of Msh2 [309]. Furthermore, the embryonic stem (ES) cells from these

mice did not demonstrate the MSI phenotype [310], implicating functions other than the post-

replicative error repair. Subsequent studies performed using an isogenic cell line (the human

embryonic cell line HEK293T) with inducible MLH1 has shown that while post-replicative error

repair can occur even when as little as 10% of MLH1 is expressed, DNA damage signaling in

response to MNNG requires full expression of MLH1[311].

Temozolomide is an antitumour triazene compound that under physiologic conditions is rapidly

metabolized to form a methylating species that reacts with DNA to form methyl adducts. The O6

MeG lesions is thought to be the key lesion by which temozolomide exerts its toxic effects [312].

However, this adduct is subject to a single-step repair reaction that simply transfers the methyl

group to a cysteine residue within the repair protein O6-methylguanine-DNA methyl transferase

(MGMT), thus restoring the DNA to its original state. Thus, MGMT is a major determinant in

temozolomide toxicity [312], but the MMR proteins have also been shown to be critical in this

process [313, 314].

Page 66: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

49

Figure 1.4 A simplified model of the MMR protein dependent DNA damage signaling

network

MMR proteins recognize and bind to damaged DNA, and then recruit other DNA damage

signaling kinases, including ATM, ATR and their effectors checkpoint kinases CHK2/CHK1.

These kinases are able to bind to and stabilize p53. The p38 mitogen-activated protein (MAP)

kinase interacts with MMR proteins and connects them with p53 and its homolog p73 another

transducer of MMR dependent damage response. MMR protein, MLH1 can activate the tyrosine

kinase c-Abl, which is then able to phosphorylate or stabilize p73. PMS2 is able to directly

interact with p73 and stabilize it. All these pathways are able to lead to cell cycle arrest and/or

apoptosis [294].

Page 67: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

50

1.3.5 Other roles of mismatch repair proteins

MMR proteins have been implicated in a number of additional process involving recombination,

meiosis and somatic hypermutation. However, these processes are not as well studied as the

post-replicative error repair function and many of the other proteins that cooperate with MMR

proteins in these functions remain to be elucidated. Several of these processes will be briefly

addressed below.

1.3.5.1 Roles of MMR in recombination

Mismatch correction in recombination intermediates

In eukaryotes, the repair of mismatches in recombination intermediates results in the

phenomenon of gene conversion[315], a process by which information of one chromosome is

replaced with information from the homologous chromosome. In the absence of functional

MMR, mismatches in recombination intermediates are not repaired. Thus, the mismatch

containing strands segregate during the first mitotic division after meiosis, a process referred to

as post-meiotic segregation. In yeast with mutant msh2, mlh1 or pms1, a decrease in gene

conversion events is observed, accompanied by a concomitant increase in post-meiotic

segregation[316-318]. Studies to date indicate that the same basic MMR machinery that detects

replication errors is also responsible for detecting mismatches in recombination intermediates.

However, it is likely that there are additional proteins that are unique to each process [253].

Regulation of recombination between nonidentical sequences

MMR has also been to shown to inhibit the homologous recombination of non-identical strands.

In Saccharomyces cerevisiae the MMR machinery can inhibit recombination between diverged

sequences both in meiosis and mitosis. Furthermore, msh2 and mlh1 mutants appears to play

Page 68: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

51

nonequivalent roles is this process, with msh2 mutants demonstrating higher rates of

recombination between diverged sequences compared to mlh1 or pms1 mutants[319, 320]. This

inhibition of crossover of diverged sequences has been proposed to be important in establishing

and maintaining genetic barriers between species[321]. Furthermore, this activity has been

proposed to promote genome stability by inhibiting interactions between diverged repetitive

DNA sequences during mitosis and meiosis [253]. By preventing these mismatched strands from

recombining, it increases the fidelity of these strand exchanges during recombination by

increasing the likelihood of interaction of two homologous fragments [274].

1.3.5.2 Roles of MMR proteins in meiosis

Yeast MSH4 and MSH5 were the first identified examples of eukaryotic MutS homologs that

have functionally diverged to carry out novel meiosis specific roles and in the process lost their

ability to repair mismatches [322]. These proteins have been proposed to directly bind Holliday

junctions and promote meiotic crossing over. Furthermore, MSH4-MSH5 proteins have been

show to interact with the MLH1-MLH3 heterodimer. Consistent with its role in meiosis, MSH4

and MSH5 expression is highest in the ovaries and testis of mammals [269, 323]. Msh5-/-

mice

are viable, but sterile [268]. Interestingly, mutation of MutL homologs appears to have a more

severe impact on the meiotic functions of mouse models. This is supported by the fact that both

male and female Mlh1-/-

mice are sterile [324, 325]. Examination of the testicular tissue from

these mice has indicated that the spermatocytes are arrested in meiosis I. Additionally, male

Pms2-/-

mice are sterile, while the corresponding females are fertile [326]. However, the cause

behind this sex-specific difference is not clear.

Page 69: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

52

1.3.5.3 Somatic hypermutation of immunoglobulins

Somatic hypermutation of the immunoglobulin (Ig) variable genes is a process that generates

high-affinity antibodies in response to antigen stimulation. It is a process that allows a small

amount of genetic material to be used to generate diverse antibodies, sufficient to deal with the

multitude of antigens [327]. The germline encodes IgM antibodies that are composed of the

variable (V), diversity (D), and joining (J) elements. However, these antibodies are not able to

mount an effective response against pathogens. Once an antigen appears and stimulates B cells,

they proliferate, differentiate and migrates to the germinal centers in secondary lymphoid organs

and become centroblasts. In these germinal centers, centroblast B cells express large amounts of

activation-induced deaminase (AID), which initiates the somatic hypermutation of the antibody

V regions (corresponds to the antigen binding sites)[328, 329]. This is thought to be the first

phase of this process, whereas the second step refers to the error prone repair of AID-induced

mutations by BER and MMR [330]. It has been proposed that in germinal centers MMR

enzymes, which normally act to maintain genomic stability, become error prone and greatly

increase the number of mutations in the V region. The role of MMR in this process is debated,

with some studies showing that a functional MMR system is required for full levels of

hypermutation [331, 332], while other studies have demonstrated that it is not the frequency but

the spectrum of hypermutation that is altered in MMR deficient mice [333].

1.3.6 The structure and expression of the MMR protein MLH1

The MutL proteins, including MLH1 are ATPases of the GHKL (gyrase/Hsp90/histidine-

kinase/MutL) family [334] with the ATPase situated in the N-terminus and the MutL

dimerization domain located in the C-terminal domain. MLH1 is located on chromosome 3p21

and is composed of 756 amino acids that are distributed over 19 exons. Additionally, MLH1

Page 70: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

53

transcript and protein levels remain constitutively expressed throughout cell cycle progression

[335].

Previous studies in mice have shown that both MLH1 and PMS2 proteins have both nuclear

localization sequences (NLS) and nuclear export sequences. However, it was found that

dimerization of MLH1 with PMS2 in the cytoplasm is required to unmask the NLS, which then

facilitates the nuclear import of the MutLα heterodimer [336]. The same mechanism may be true

in the human system, where studies have recently identified two putative NLS in MLH1 and one

candidate NLS in PMS2 [337].

Moreover, MLH1 is also a component of the BRCA1-associated genome surveillance complex

(BASC), which consists of proteins such as BRCA1, MSH2, MSH6, ATM, BLM and the

RAD50-MRE11-NBS1 protein complex [338]. All these proteins have a role in recognizing

abnormal DNA structures or damaged DNA, thus BASC may serve as a sensor for DNA damage

[338].

The MLH1 promoter region is arbitrarily divided into four regions (A- D) with respect to its

translation start site; A (-711 to -577), B (-552 to -266), C (-248 to -178) and D (-109 to +15)

[339]. While hypermethylation of the entire [A+B+C+D] region is associated with gene silencing

and loss of MLH1 expression, this correlation is augmented when the proximal region of the

MLH1 promoter is hypermethylated [340]. Methylation of the C+D region of MLH1 is invariably

linked with gene silencing [339, 341], suggesting this to be the core promoter region of this gene.

Recent evidence indicates that the promoter of MLH1 is bi-directional with a second gene,

EPM2AIP1 located in a head-to-head orientation on the opposite strand 321 base pairs away

from the MLH1 transcription start site [342]. The ubiquitously expressed EPM2AIP1 gene is

comprised of a single large exon, 1824 nucleotides in length encoding a 607 amino acid protein

Page 71: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

54

with no known homology to other proteins or structural motifs [343]. The function of

EPM2AIP1 is currently not known.

1.3.7 Mismatch repair and disease

Mutations in MLH1 [344, 345] and MSH2 [346, 347] account for the more than 80% of known

cases of Lynch Syndrome. Other MMR genes implicated to date include MSH6, PMS2, and

possibly MLH3. MMR deficiency has also been shown to give rise to ~15% of sporadic

colorectal, endometrial and gastric cancers. The majority of such sporadic CRCs associated with

microsatellite instability are primarily due to epigenetic inactivation of MLH1 due to promoter

hypermethylation [121, 122].

Germline MMR defects also lead to Muir-Torre syndrome (MIM #158320). Muir-Torre is

characterized by tumours in the sebaceous glands or keratoacanthomas that are associated with

one or more of the various visceral neoplasms, mainly colorectal, endometrial, urological, and

upper gastrointestinal neoplasms. It has been proposed that Muir-Torre is a clinical variant of the

Lynch Syndrome [348]. However, about 35% of tumors in individuals positive for the Muir-

Torre Syndrome do not display instability of microsatellite repeats, which combined with other

features of Muir-Torre has spurred the speculation that at least two distinct forms of Muir-Torre

exist[349]. The first sub-type, which includes MSI positive tumors, shares genetic and

pathological features with Lynch syndrome. It is characterized by early-onset colorectal

carcinoma and a strong family history of cancer. The second group of MSS or MMR proficient

tumors shows later ages of onset and a less pronounced family history, and is likely due to genes

that are not implicated in MMR pathway. Muir-Torre syndrome is associated with germline

mutations in the MSH2 gene, and to a lesser extent with mutations in MLH1 and MSH6 genes

[350-353]. We recently reported a family where a complex MSH2 mutation predisposed to both

Page 72: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

55

Lynch and Muir-Torre syndromes [354]. Interestingly, a subset of tumours in this family

displayed neither prototypical feature of microsatellite instability or immunohistochemistry,

highlighting the variable phenotypic manifestations associated with genetic alterations of the

MMR genes.

Furthermore, there are also reports of individuals who are homozygous for mutations in the

mismatch repair genes MLH1, MSH2, MSH6 and PMS2. Such individuals develop several

congenital abnormalities including haematopoietic malignancies, pediatric brain cancers,

childhood leukemia, HNPCC-related cancers and multiple café-au-lait spots, a common

characteristic of neurofibromatosis type 1(MIM#162200) [355-360]. This phenotype manifests in

an autosomal recessive fashion, because a mutant allele is inherited from each parent.

Interestingly, the presence of early-onset gastrointestinal cancers along with the features of

neurofibromatosis type 1 (NF1) was first reported in children who were positive for a

homozygous missense variant in exon 18 of MLH1 (Arg687Trp) [356]. These children had

inherited each variant allele from their parents, who were heterozygous carriers of this variant.

In addition, mutations in both APC as well as the MMR genes, MLH1 and PMS2 are associated

with Turcot‟s syndrome (MIM#276300) [361]. There have also been reports of individuals

carrying compound heterozygous PMS2 mutations who develop Turcot‟s syndrome [362]. This

syndrome is defined by the presence of brain tumours and multiple adenomas/colorectal cancers

that occur at an early age. The central nervous system tumours in individuals with APC

mutations are typically medulloblastoma, whereas those with MMR gene mutations are usually

glioblastoma multiforme.

Page 73: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

56

1.4 Genetic Variation in Mismatch Repair

1.4.1 Predictive models of germline variants in MMR genes

The ability to predict germline MMR mutations would be useful in situations where the existence

of an inherited defect cannot be directly addressed in the laboratory. Furthermore, universal

germline testing of all index CRC cases is not feasible given that family histories of CRC is

common, and that mutations that lead to this syndrome are both rare and scattered across several

genes[363]. Several groups have attempted to translate the extensive understanding of Lynch

syndrome into clinically useful prediction algorithms, similar to those developed for breast[364],

melanoma[365] and pancreatic syndromes[366].

The Leiden (Wijnen) model was developed to address this void in Lynch syndrome [367]. This

model estimates the probability of carrying a mutation in an MMR gene based on mean age of

CRC diagnoses, the presence of endometrial cancers in family members and the fulfillment of

Amsterdam criteria. Despite its utility, the Leiden model does not use all available information

and as well does not provide risk predictions.

Currently there are several computer-based models; MMRpro [368], prediction of mutations in

MLH1 and MSH2 (PREMM1,2)[369] and MMR predict[370], which estimate the probability that

an individual carries a mutation in a MMR gene. MMRpro is a model that estimates the

probability that an individual carries a deleterious mutation in an MMR genes, MLH1, MSH2,

and MSH6. Data for MMRpro was obtained by meta-analysis of population-based studies. It

also incorporates information from published cases along with data on the specificity and

sensitivity of MSI status and germline testing. This probability is evaluated using a detailed

family history of CRC and endometrial cancer, which includes information for the individual

Page 74: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

57

being counseled and each of his or her first- and second-degree relatives. Furthermore, MMRpro

estimates future cancer risks for unaffected individuals (mutation positive or negative), as well as

untested individuals. PREMM1,2 determines the likelihood that an individual carries a mutation

in MLH1 and MSH2. This model used specific information about an individual‟s age, sex, as

well as specific details about the individual‟s family history as supplied on the test submission

forms at Myriad Genetics Inc., a genetic testing laboratory. MMRpredict calculates the

probability than an individual carries a mutation in either MLH1 or MSH2. Data was obtained by

determining the germline mutation status of 870 individuals followed by MSI and IHC testing.

The presence of germline mutation was correlated with variables such as age, sex, family history,

and presence of endometrial cancer to determine the weight of each predictor. Following this,

information regarding MSI and IHC was incorporated into the equation.

In an independent validation study carried out in a cancer genetics clinic using a high-risk cohort,

all three models performed similarly [371]. In an independent study of families with a

moderately high risk of developing CRC, all three programs performed comparably, while the

PREMM1,2 model was found to demonstrate the best performance [363]. In fact in this study,

these models performed better than Amsterdam II criteria leading the authors to propose that

these models may soon replace the Amsterdam and Bethesda criteria. Unlike Amsterdam or

Bethesda criteria, which are only able to identify the existence of risk, these programs are able to

provide an estimation of risk. This provides the opportunity to set up probability thresholds for

interventions that will assist with clinical decision making [363]. However, these cut-offs remain

to be determined. It is expected that the ability to predict the presence of germline mutations will

result in an increased rate of mutation discovery in the near future.

Page 75: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

58

1.4.2 Spectrum of genetic alterations in the major MMR genes

Genetic alterations in the MMR genes range from large genomic deletions and duplications,

truncating or missense variants to splice site alterations, which occur in distinct domains of

MLH1 or MSH2. A recent database catalogues over 1500 MMR gene variants

(http://www.med.mun.ca/MMRvariants) [372]; while the International Society for

Gastrointestinal Hereditary Tumors (InSiGHT, http://www.insight-group.org/) lists over 400

pathogenic mutations that have been identified in the MMR genes. MMR genes act as tumour

suppressors, in that one wild-type allele is sufficient to maintain normal function. Individuals

with Lynch syndrome have only one allele that is inactivated (the one bearing the germline

mutation). Thus, for the process of carcinogenesis to proceed an independent somatic event must

occur on the wild-type allele rendering the MMR gene inactive.

More recently, in a subset of Lynch syndrome cases constitutional epimutations of the MLH1

gene were identified[373]. These epimutations are characterized by soma-wide methylation of a

single allele of the promoter and allelic transcriptional silencing. Germline MLH1 epimutations

are functionally equivalent to an inactivating mutation and have been shown to produce a clinical

phenotype that resembles Lynch syndrome. However the inheritance of epimutations was found

to be weak, thus limiting the utility of family history in guiding screening. In addition MLH1

expression is altered by several epigenetic mechanisms. For example, the promoter region of

MLH1 is hypermethylated in the majority of MSI-H sporadic cancers. Furthermore, another

study found that miR-31 and miR-223 were overexpressed in colorectal tumours of patients with

Lynch syndrome, indicating the potential role for miRs in regulating the expression of genes

involved in susceptibility to Lynch syndrome [374].

Page 76: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

59

A recent study offers another novel mechanism of Lynch syndrome susceptibility. Here

heterozygous germline deletions of the last exons of TACSTD1 (encodes EPCAM); a gene

directly upstream of MSH2 led to the somatic silencing of the corresponding MSH2 allele by

hypermethylation. [375]. Findings from another study indicate that while deletions of EPCAM

account for only a small proportion of individuals suspected of Lynch syndrome, these deletions

were more frequent in individuals with genetically confirmed Lynch syndrome [376]. A recent

report described somatic hypermethylation of MSH2 in panel of a Lynch syndrome tumours that

were deficient for MSH2 expression [377]. In contrast MSH2 hypermethylation was not

observed in normal colonic tissues or sporadic CRCs (both MSI/MSS). While a subset of these

tumours had deletions in EPCAM, the majority were not associated with defects in EPCAM but

instead were positive for germline mutations in MSH2 [377]. Although further studies are needed

to confirm this finding, it suggests a unique mechanism for MSH2 inactivating the second, wild-

type allele of MSH2.

About 16% of all MSH2 and 30% of all MLH1 alterations are missense variants of no obvious

clinical significance [378]. Germline MSH2 mutations are associated with loss of MSH2 and/or

MSH6 expression in the majority of cases. Consequently, a germline MSH2 missense variant in

patients presenting with a family history of CRC and a MSH2 deficient CRC tumour is likely to

be a pathogenic mutation. On the contrary the significance of the majority of missense variants

identified in MLH1 is largely unknown. This issue is compounded by the fact that genetic

alterations are dispersed across the gene, with no known hot spot regions associated with

confirmed disease risk. Given that the diagnosis of Lynch syndrome relies primarily on the

identification of germline defects in the MMR genes [234], detecting these unclassified genetic

variants creates much ambiguity in the clinical setting, as the pathogenicity of these variants

cannot be readily ascertained. This affects the accuracy of predictive genetic testing and

Page 77: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

60

counseling of patients carrying these variants, as well as their family members at risk of

developing Lynch syndrome.

1.4.3 Approaches to classifying unclassified variants

Several different strategies are employed to characterize the pathogenic and functional

significance of MMR variants, each with specific strengths and limitations [234, 379-385].

Genetic linkage assessment is based on the co-segregation of a MMR variant with disease status

in pedigrees, which is relatively straightforward to assess. However, it requires screening of

multiple affected individuals in the same family, which may not always be feasible with smaller

families or when clinical specimens are unavailable for all affected members in a family. MMR

variants that occur concurrently with known deleterious mutations could potentially act

synergistically with the deleterious mutation or have no effect on the disease phenotype. Thus, in

this case a less pathogenic or benign nature is indicated, although there is no direct evidence for

the lack of pathogenicity of such a variant. In addition, genetic epidemiologic studies can provide

direct evidence of association with cancer risk. However, if these studies are to be informative a

prohibitively large sample population is required, particularly for variants that occur with low

frequency. Bioinformatics approaches that rely on different databases (e.g. SIFT or PolyPhen)

are increasingly being used to predict the potential impact of a variant on MMR gene structure

and/ or function(s). The advantages of these approaches are that detailed family histories or large

patient populations are not required. However, their predictive value is limited unless a panel of

such programs are used [386]. Several groups have attempted to develop functional assays based

on the sequence homology between human and yeast MLH1[387]. One assay defined variants

that were unable to restore genetic stability in a mlh1 deleted yeast host strain as pathogenic.

Another assay made use of the finding that overexpression of mlh1 in yeast led to defective

Page 78: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

61

MMR. Based on the finding that wild-type MLH1 behaved in a dominant negative fashion and

led to MMR defects, any variant that did not result in defective MMR in yeast was deemed to be

pathogenic [384]. However, the utility of these assays depend upon the homology between yeast

and human sequences, which is poor in the central region of the MLH1 protein. Guerrete et al.,

described a GST fusion assay where a radiolabelled variant of MLH1 was assessed for its ability

to interact with PMS2 [388]. However this method cannot be used to evaluate variants that do

not play a role in heterodimerization. Functional and biochemical studies carried out in human

systems provide the most direct evidence because they are able to assess the effects of genetic

variants on protein stability, expression and function(s). For this approach alterations are created

by site-directed mutagenesis in plasmids containing the wild-type gene, which are then used to

assay expression, MMR function or protein stability [383, 385, 389-391]. The ability of missense

variants to alter the efficiency of the post-replicative error repair can be assessed using several

mismatch repair assays. One prototypic approach relies on the ability of variant MLH1 protein to

correct a mismatch that occurs in a DNA heteroduplex [392]. Briefly, the G•T mispair renders

this heteroduplex resistant to cleavage by restriction enzymes; however if post-replicative error

repair is intact then this mismatch is repaired to an A•T. This renders it sensitive to cleavage,

making it possible to evaluate repair efficiency by measuring cleavage efficiency. Nonetheless,

these assays can only determine a variant‟s ability to alter the post-replicative error repair

function. Therefore, at present it remains important to use a combination of the above approaches

to accurately assess the pathogenicity associated with a variant. However, determining the most

appropriate assays requires a better understanding of the regulation and function of MLH1, as

well as the role of missense variants in affecting these processes. Furthermore, as our

understanding of the role of MLH1 and the MMR system evolves, it is remains important to

devise new assays that take this information into consideration.

Page 79: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

62

1.5 Hypothesis and Objectives

I hypothesize that missense alterations in distinct domains of MLH1 likely affect its expression

and function(s) to varying degrees.

In order to characterize the possible effects of unclassified variants on MLH1 and the MMR

system, the following objectives were formulated:

1. Assess the effect of unclassified variants on MLH1 expression, stability and function

2. Determine if MLH1 genetic variants lead to unbalanced allelic expression

3. Investigate the functional effects of the MLH1 c.-93G>A variant on MLH1/EPM2AIP1

promoter activity

Page 80: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

63

CHAPTER 2

The role of unclassified variants in influencing MLH1 expression, stability

and function

Sheron Perera and Bharati Bapat

Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada

Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, Canada

Department of Pathology and Laboratory Medicine, Mt. Sinai Hospital, Toronto, Canada

All work in this chapter and data analysis was carried out by Sheron Perera, with the exception

that Vaijayanti Pethe assisted with the generation of K618A stable inducible cell line.

The majority of the data presented in this chapter has been previously published in Human

Mutation: Perera S., and Bapat B. (2008). The MLH1 variants, Arg265Cys and Lys618Ala affect

protein stability, while Leu749Gln affects heterodimer formation. Human Mutation 29(2):332.

Reproduced with permission from John Wiley and Sons, copyright 2008.

Page 81: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

64

2 Chapter 2 The role of unclassified variants in influencing

MLH1 expression, stability and function

2.1 Summary

Germline mutations in the mismatch repair genes lead to the Lynch syndrome/ HNPCC, and

genetic testing is offered to identify individuals at risk of developing this disease. About a third

of all genetic alterations detected in the mismatch repair gene MLH1 are missense variants, the

majority of which are of equivocal clinical significance. In this report, we investigate the

molecular basis of the pathogenicity of four missense variants localized to distinct functional

domains. Our results demonstrate that the MLH1 variants p.Arg265Cys and p.Lys618Ala

significantly decrease the stability of the protein, while another missense variant p.Leu749Gln is

able to compromise heterodimerization of the MLH1-PMS2 complex. Furthermore, using a

stable inducible cell line model we were not able to assess the impact of these substitutions on

altering MLH1 function likely due to the disruptive effects associated with MLH1

overexpression in cells.

2.2 Introduction

Lynch syndrome or hereditary nonpolyposis colorectal cancer (HNPCC), is a common cancer

predisposition syndrome that is inherited in an autosomal dominant manner. Lynch syndrome is

associated with germline mutations in the mismatch repair (MMR) genes. Individuals in Lynch

syndrome families are at an increased risk of developing colorectal cancer (CRC), while women

in these kindreds are predisposed to endometrial cancer. In the clinical setting, diagnosis of

Lynch syndrome is complicated by the lack of precise characteristic features. While clinical

guidelines including Amsterdam Criteria 1 and 2, and Bethesda Criteria have proven to be useful

Page 82: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

65

in diagnosis, these criteria are limited in their ability to accurately identify patients with Lynch

syndrome. Thus, the diagnosis of Lynch syndrome relies primarily on the detection of germline

defects in the MMR genes of these patients [234].

The MMR pathway plays a central role in maintaining genomic stability by repairing both

mismatches and insertion/deletion loops that arise during DNA synthesis [393]. MMR improves

the fidelity of DNA biosynthesis several hundred-fold and reduces the error rate to about one

error per 1010

bases [279]. Defective MMR results in the mutator phenotype, which increases the

likelihood of mutations in a battery of genes that regulate growth, differentiation or apoptosis.

Deficiencies in MMR lead to microsatellite instability (MSI). Tumours from the majority of

Lynch syndrome patients (>85%) display MSI, making it a hallmark feature of this syndrome

[123, 124].

In addition to the error repair function, MMR proteins have been implicated in signaling DNA

damage and apoptosis [293]. Recognition and perhaps processing of these lesions by MMR

proteins leads to activation of DNA damage signaling pathways, which can in turn result in cell-

cycle arrest or apoptosis. Although the role of the individual MMR proteins in post-replicative

DNA repair is relatively well defined, their contributions to signaling DNA damage is not as well

understood. Emerging research indicates that unlike many of the other DNA repair pathways

that mediate resistance to DNA damage, MMR is necessary to mediate the toxicity of several

anti-cancer drugs [394]. Therefore, a better understanding of how the MMR proteins signal DNA

damage will help us to better understand the mechanism by which MMR deficient tumours are

refractory to certain classes of chemotherapeutic agents. Interestingly, previous studies with

MMR mouse models have demonstrated that missense variants are capable of separating the

mismatch-error repair function from the DNA-damage response function of the MMR system

Page 83: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

66

[395]. This underscores the utility of missense alterations as biological tools capable of providing

valuable insights into the role of MMR proteins in signaling DNA damage [380, 396].

Advances in the understanding of the genetic basis of Lynch syndrome has facilitated predictive

genetic testing programs to identify individuals with an inherited MMR mutation who are at an

increased risk of developing CRC and other Lynch syndrome -associated cancers. By predicting

risk prior to the progression of disease, these programs play an integral role in the effort to

improve surveillance, treatment and prevention of cancer. A recent database catalogues over

1500 MMR gene variants (http://www.med.mun.ca/MMRvariants [372]); while the International

Society for Gastrointestinal Hereditary Tumours (InSiGHT, http://www.insight-group.org/) lists

over 400 pathogenic mutations that have been identified in the MMR genes. Among these,

mutations in MLH1 and MSH2 are responsible for over 85% of known cases of Lynch syndrome

[397]. About 25% of MLH1 alterations are missense alterations of equivocal clinical significance

[378]. Detection of such an unclassified variant affects the accuracy of predictive genetic testing

and counseling of patients carrying these variants, as well as their family members at risk of

developing Lynch syndrome. It is likely that missense alterations of MLH1 affect its expression

and function(s) to varying degrees, thereby contributing to the variable disease phenotype

observed in Lynch syndrome.

In this study we investigate the effects of four representative missense variants in MLH1 on its

expression and function, in order to clarify their pathogenic significance (Figure 2.1). We

functionally characterized the MLH1 alterations c. 55A>T leading to the substitution Ile19Phe,

c.793C>T leading to Arg265Cys, both of which occur adjacent to the ATPase domains. We also

studied c.1852_1853AA>GC leading to Lys618Ala, which is located in the MutL homolog

binding domain. The c.55A>T variant has been reported previously however, no functional

Page 84: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

67

studies have been carried out to date [398, 399].The pathogenicity of the R265C and K618A

variants remain debatable even after several analyses due to the existence of conflicting data

[234, 385, 388, 400-404]. We also hypothesized that missense alterations that occur in the C-

terminal region, adjacent to the MutL homolog binding domain likely affects MLH1

heterodimerization, and to investigate this we constructed an alteration c.2246T>A which leads

to the missense substitution Leu749Gln and is located within the c-terminal homology domain.

Page 85: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

68

Figure 2.1 Schematic representation of the functional domains identified in MLH1.

Arrows indicate the locations of the representative variants selected in this study.

I19F

Page 86: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

69

2.3 Materials and Methods

2.3.1 Materials and cell culture

The human colon cancer cell line, HCT116 cell line was obtained from American Type Culture

Collection (Rockville, MD) was maintained in McCoy‟s 5A medium supplemented with 10%

FBS. The HCT116 cell line with the additional copy of chromosome 3 was a kind gift from Drs.

Minoru Koi and Richard Boland (University of Michigan Medical School, Anne Arbor, MI). The

cell line was developed by transferring a single copy of human chromosome 3 from fibroblasts to

HCT116 cells via microcell fusion [405]. These cells were maintained in McCoy‟s medium with

10% FBS and 400μg/ml of G418 (Sigma). The vectors containing the full-length wild-type

MLH1 cDNA (GenBank accession no. NM_000249.2) and PMS2 cDNA (NM_000535) was

kindly provided to us by Dr. G. Plotz and Dr. R. Kolodner. Benzylguanine and temozolomide

(TMZ) were purchased from Sigma Chemicals dissolved in DMSO, and stored at -80°C in single

use aliquots.

2.3.2 Identification of MLH1 variants

The variants I19F, R265C and K618A were identified in patients that were screened through

the Ontario Predictive Genetic Testing Program at Mt. Sinai Hospital, Toronto, Canada. Genetic

screening is initiated by performing MSI testing, and immunohistochemistry (IHC) for the MMR

proteins on tumour specimens of individuals presenting with either a family history of CRC,

and/or those who develop tumours at a young age of onset (<50). This is followed by multiplex

ligation-dependent probe amplification (MLPA), to detect large genomic deletions and

duplications in the MMR genes. If such changes are not detected, germline DNA is screened by

sequencing the MLH1 and MSH2 genes, guided by the MSI/IHC results of patient tumours.

Page 87: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

70

Through this program we have identified many established germline mutations, known to cause

aberrant expression of protein, as well as several unclassified variants.

2.3.3 Computational Assessment of Variants

We used the computational programs Polymorphism Phenotyping (PolyPhen)

(http://genetics.bwh.harvard.edu/pph/), and Sorting Intolerant From Tolerant (SIFT)

(http://blocks.fhcrc.org/sift/SIFT.html) to predict the pathogenicity of these variants. PolyPhen

uses structural information, along with multiple sequence alignments to classify the

pathogenicity of an amino acid substitution with an accuracy of about 80% [406]. Based on these

criteria it predicts a substitution to be benign, possibly pathogenic or probably pathogenic

respectively, in ascending order of its predicted impact. SIFT uses an algorithm that aligns

closely related sequences, and calculates normalized probabilities for all possible substitutions

[407]. It has been reported that SIFT is able to predict the impact of non-synonymous

substitutions with an accuracy ranging from 65-92% in a range of genes associated with human

disease [386]. It predicts positions with normalized probabilities of less that .05 as deleterious,

while those with probabilities greater than or equal to .05 are classified as tolerated. Default

settings were used for both programs.

2.3.4 Site-directed mutagenesis and construction of MLH1 mutants

We used site-directed mutagenesis to create the expression constructs according to the

specifications of the manufacturer (Quick Change Site-directed Mutagenesis, Stratagene,

LaJolla, CA). MLH1 cDNA coding for the variants were generated using primer pairs that

contained the specific nucleotide alteration (Table 2.1). To confirm the integrity and the presence

of the respective missense substitutions, all cDNA inserts were directly sequenced. We sub-

Page 88: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

71

cloned these constructs into myc-His tagged expression vectors (Invitrogen, Carlsbad, CA), to

use them for a wider range of experimental procedures. All mutation numbering is based on

cDNA sequence, with codon 1 corresponding to the major initiation codon. Primer sequences and

PCR conditions for all protocols listed above are available on request.

2.3.5 RNA extraction, cDNA synthesis and sequencing

RNA extraction was performed using TRIzol according to the manufacturer‟s protocol

(Invitrogen Life Technologies, Burlington, ON). Reverse transcription PCR was performed

according to standard techniques as described before [408]. In brief, cDNA was generated from

total RNA by use of random hexamers pd(N6) (Amersham Pharmacia, Piscataway, NJ), oligo dT

(Invitrogen Life Technologies), 1 x first strand buffer, 0.5 mmol/l of deoxynucleotide

triphosphate, 5 mmol/l DTT, and 200 units of SuperScript II (Invitrogen Life Technologies).

cDNA was generated by incubating at 45°C for one hour and 70°C for 15 minutes.

PCR was

carried out to amplify the region surrounding the nonsense mutation on MLH1. Following the

PCR reaction, DNA was isolated by gel electrophoresis and purified by QIAquick Gel Extraction

Kit (Qiagen, Mississauga, ON, Canada). Sequencing reactions were carried out using an ABI377

sequence analyzer according to the manufacturer‟s protocol(Applied Biosystems, Foster City,

CA).

2.3.6 Production of recombinant proteins and western blotting

HCT116 cells were transfected using Lipofectamine 2000 reagent (Invitrogen), according to the

manufacturer‟s instructions with the appropriate vectors and harvested at the indicated time

points. For total protein extraction, cells were washed twice with cold phosphate buffered saline

(PBS). Cold radioimmunoprecipitation assay (RIPA) lysis buffer (0.5 % Nonidet P-40, 0.5%

Page 89: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

72

sodium deoxycholate, 0.1% sodium dodecyl sulfate, 1x PBS) buffer containing Complete

protease inhibitor tablets (Roche, Mannheim, Germany) was used to lyse the cells on ice for

approximately 20 minutes. The lysates were then collected by centrifugation at 14,000 rpm for

approximately 15 minutes. The expression levels for the proteins were examined by sodium

dodecyl sulphate/ polyacrylamide gel electrophoresis, followed by western blot analysis with

anti-MLH1 (clone 168-728; BD Biosciences) and anti-post meiotic segregation increased 2

(PMS2; MIM# 600259)) (clone A16-4; BD Biosciences) antibodies. β-Actin antibody (Sigma)

was used to assess equal loading in all lanes. To validate our results, we co-transfected cells with

0.15 g of pCMV β-galactosidase vector, and measured its enzymatic activity as described

previously [409].

2.3.7 Combined coimmunoprecipitation and western blot analysis

For coimmunoprecipitation 1000 μg of whole cell lysate transfected with each of the constructs

were incubated with 2.5 μg of anti-MLH1 antibody. The lysates were then incubated for a

further 1.5 hours after the addition of 40 l of protein A/G agarose beads (Santa Cruz

Biotechnology, Santa Cruz, CA). The precipitates were then centrifuged at 1000 rpm for 2

minutes, and washed 3 times with 500 l of RIPA buffer and resolved on a 10% SDS-PAGE gel.

Interactions between MLH1 and PMS2 proteins were detected by western blot analysis with anti-

MLH1 and anti-PMS2 antibodies.

2.3.8 Pulse chase assays

HCT116 cells were plated in 6 cm dishes, and then transiently transfected with wild-type and

variant MLH1 cDNA. 24 hours post transfection, the media was aspirated from the plates and the

cells were washed twice in warm PBS. 1 ml Methionine (Met) -, and Cysteine (Cys)-free

Page 90: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

73

DMEM (Sigma - supplemented with Glutamine) was added to the cells, and incubated for 30

minutes at 37 C to starve the cells of endogenous pools of Met and Cys. Thereafter 150μCi of

EasyTag Express 35

S (Perkin-Elmer, Waltham, MA) was added to each plate, and the cells were

incubated for an additional 40 minutes at 37 C. The radioactive media was then removed and the

cells were washed twice with warm PBS. For the chase, 5 ml of warm DMEM supplemented

with 2mM Methionine and 2mM Cysteine was added to all plates except the „0‟ time point. The

plates were then incubated at 37 c and harvested at the specified time points from 0-30h. The

harvested lysates were pre-cleared for one hour with protein A/G agarose beads. Thereafter, first

total MLH1, and then total β-Actin was immunoprecipitated, and analyzed by SDS-PAGE

followed by autoradiography.

2.3.9 Cycloheximide experiments

HCT116 cells were transfected with either wild-type or variant MLH1 constructs. 24 hours post-

transfection, cells were treated with 25μg/ml of cycloheximide (Sigma) or vehicle (DMSO), and

lysed at indicated time points as described before. The levels of MLH1 were assessed by western

blotting.

2.3.10 Generation of stable inducible cell lines

A tetracycline-regulated expression system (T-rex) was used to generate HCT116 cell lines with

inducible expression of wild-type or variant MLH1 according to the manufacturer‟s instructions

The T-Rex system is a tetracycline regulated mammalian expression system that uses elements

from the E.coli TN10-ecoded tetracycline operon. In the absence of tetracycline or doxycycline

(a tetracycline derivative with a longer half life), the Tet repressor (TetR) binds the Tet operator

Page 91: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

74

sequences in the inducible expression vector, repressing the transcription of wt or variant MLH1.

When added into the medium tetracycline (or doxycycline) binds the Tet Repressor, resulting in

its release from the Tet operator sequences, which results in the induction of wt or variant MLH1

expression.

To generate an HCT116 cell line with stable expression of TetR, HCT116 cells were seeded in 6

cm plates. 24 hours later cells were transfected with 10µg of pcDNA6/TR per plate using

Lipofectamine 2000 (Invitrogen Life technologies) transfection reagent according the

manufacturer‟s instructions. 48 hours post-transfection cells were re-seeded into 15 cm plates at

low density and grown in the presence of 5µg/ml of blasticidin (Invivogen, San Diego, CA) to

select for transfectants. Colonies that were established were carefully isolated, expanded and

analyzed by western blotting using the TetR antibody (Invitrogen life technologies).

To generate the doxycycline inducible 2xmyc-tagged MLH1/pcDNA4/TO construct, the 2xmyc-

MLH1/pcDNA3.1 construct was digested with ApaI and EcoRV. The fragment encoding the

MLH1 was subcloned into the pcDNA4/TO inducible expression vector, which was also

digested with ApaI and EcoRV. The constructs for I19F, K618A were also generated in a similar

manner.

To generate stable inducible clones, the HCT116-TetR cells were transfected with 2xmyc-

MLH1/pcDNA4/TO as described above and transfectants were selected for in the presence of

both 5µg/ml blasticidin and 100µg/ml zeocin (Invivogen). Zeocin resistant colonies were

selected and analyzed by western blotting using the myc-tag antibody (9e10, Santa Cruz

Biotechnology). A similar protocol was followed to generate HCT116 cell lines that stably

expressed the variant MLH1 proteins. To screen resistant colonies, cells were treated with either

1 µg/ml Dox or vehicle (water). Twenty four hours later-whole cell lysates were harvested and

Page 92: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

75

analyzed by western blotting using the myc-tag antibody to detect the myc-tagged wt or variant

MLH1 proteins. Positive clones were maintained in the presence of 5µg/ml blasticidin and 100

µg/ml of zeocin.

Clones that show minimal levels of expression in the absence of treatment and maximal level of

expression upon induction were chosen and propagated. For each of the variants, we attempted to

choose the clones that show expression comparable to wild-type MLH1. Using the above process

we were able to generated three cell lines expressing wild-type MLH1, I19F MLH1, K618A

MLH1 and the empty vector. To characterize these cell lines, cells were induced with varying

doses (0.01-1μg/ml) doxycycline to determine the lowest concentration of drug that yields robust

expression of wild-type and variant MLH1.This was followed by a time course of MLH1

induction in these cell lines, where MLH1 expression was assayed 3-24 hours post-induction.

2.3.11 Cell survival assays

To assess if the missense variants in MLH1 alter cell proliferation in response to

chemotherapeutic agents, HCT116 cells expressing wild-type or variant MLH1 were treated with

temozolomide. Results generated from the empty vector (EV) cell line will help determine the

contribution of other doxycycline responsive genes to the phenotype measured. Cells were

seeded in 96-well plates at a density shown to previously to allow for exponential growth

throughout the exposure period. The next day the cells were exposed to varying concentrations

of temozolomide (0-500 µM) in the presence of 10-40 µM O6-benzylguanine over a 5 day

period. O6-benzylguanine is a inhibitor of MGMT, an enzyme that efficiently removes alkylating

damage. Thus, treatment was carried out to determine the independent contribution of the MMR

system to DNA damage signaling. Thereafter, cell survival was measured using an MTS based

Page 93: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

76

assay (CellTiter 96AQueous One Solution, Promega, Madison, WI) that measures the ability of

metabolically active cells to cleave the tetrazolium salt MTS to the water-soluble formazan.

2.3.12 Cell cycle analysis

The profiles of cell populations in G1, S and G2 phases were assessed by flow cytometry.

Briefly, cells were plated in 6 cm dishes. The next day the medium was changed to low serum

(0.5%) McCoy‟s medium and cultured for 24 hours to synchronize the phase of the cells. The

next day, cells were treated with TMZ and O6-benzylguanine, which was added to medium that

contained 10% FBS. The cells were fixed with 70% ethanol overnight at -20°c, treated with

RNaseA for 30 minutes at 37°c and stained with propidium iodide (PI). The percentage of cells

in each phase was determined using a BD FACSCalibur (BD Biosciences, San Jose, CA) and the

resulting data analyzed using the FlowJo software.

2.3.13 MSI analysis

To minimize variability that could be introduced by a mixed cell population a single clone was

selected from each stable inducible cell lines by sparsely plating cells and growing them in the

presence of blasticidin and zeocin. This clone was then expanded and grown independently for

30 generations either in the presence or absence of doxycycline (the cells were not subjected to

any treatment with alkylating agents). Cells were harvested at passage 0 (onset), 10, 30, and

chromosomal DNA was extracted using the QiaAMP DNA mini kit (Qiagen). MSI was assessed

at the mononucleotide repeat loci BAT25 and BAT26 as described before [410].

Page 94: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

77

Table 2.1 Primers used for site-directed mutagenesis

Variant Forward Primer Reverse Primer

I19F T7 F

5‟TAATACGACTCACTATAGGG3‟

ML1R

5‟ACCTCGAAAGCCATAGGTAG3‟

R265C M265F

5‟AGGATAAAACCCTAGCCTTC3‟

ML3R

5‟ATGGGTGTGTGTTTTTGGGC3‟

K618A ML7F

„AACTGTTCTACCAGATACTC‟

ML7R

„CACCTCAGTGGC TAG TCG‟

L749Q ML8F

5‟GGAGGGACTGCCTATCTTC3‟

MCSR

5‟TGATCATCGGGTTTAAACGG3‟

Table 2.2 Summary of predictions generated by computational tools

MLH1

Variant

Nucleotide

Change

Exon

Functional

Domain

SIFT22

PolyPhen23

I19F

R265C

K618A

L749Q

c.55A>T

c.793C>T

c.1852_1853

AA>GC

c.2246T>A

1

10

16

19

-

-

MutL homolog

interaction

C-terminal homology

Intolerant

Intolerant

Tolerant

Intolerant

Possibly Damaging

Probably Damaging

Possibly Damaging

Probably Damaging

Output generated by SIFT: Intolerant or Tolerant

Output generated by PolyPhen: Probably Damaging > Possibly Damaging> Benign

Page 95: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

78

2.4 Results

2.4.1 In silico analysis of MLH1 variants

For this study we chose four variants located in distinct domains of MLH1. Missense variants

that lead to the I19F amino acid substitution have been reported twice in the literature [398, 399].

Variants that lead to multiple amino acids substitutions at codon 265 have been reported in the

literature including the polymorphism R265H and the unclassified variant, R265C [385]; while

codon 618 incurs a point mutation leading to the substitution K618T, a bona fide mutation and a

two base-pair substitution leading to the variant K618A [390]. Similarly, codon 749 is shown to

result in a substitution leading to L749P [411, 412]. We first employed the computational tools,

SIFT and PolyPhen, to predict the pathogenicity of the missense substitutions chosen for this

study (Table 2.2). SIFT has been used to predict the pathogenicity of the variants R265C and

K618A in previous reports [383, 413]. However, PolyPhen is yet to be applied to the analysis of

these variants. All four of the missense variants chosen were predicted to be pathogenic by either

of these two programs. However, we observed that these computational databases did not always

provide concordant predictions. For example, while SIFT predicts K618A as a “tolerated”

alteration, PolyPhen predicts it to be possibly damaging.

To gain a better understanding of how these substitutions affect the structure and consequently

the function of the protein, we mapped this variant onto the crystal structure of the MLH1

homolog in Escherichia coli, MutL (PDB accession no.: 1B63, Ban and Yang, 1998). A high

degree of sequence conservation is seen only within the N-terminal domain of E. coli MutL (Ban

and Yang, 1998; Guarne et al., 2004). The C-terminal region of E. coli MutL shares little

sequence similarity to other MutL homologs except human MutL homolog 3 (MLH3) and PMS2

(Guarne et al., 2004). In addition, these authors were unable to detect sequence conservation

Page 96: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

79

between E. coli MutL and human MLH1 (Guarne et al., 2004). Therefore, we decided to use this

approach to assess the I19F and R265C variants that occur in the N-terminal region of MLH1.

The amino acid I19 corresponds to I14 in E.coli, and occurs on the periphery of the structure.

Based on its terminal location, it likely plays a role in mediating protein-protein interactions

(Figure 2.2). The amino acid R265 corresponds to R261 in E.coli, and it is located between an

alpha helix and a beta sheet (Figure 2.2). R261 interacts with the carbonyl groups of Ser91 and

Leu92; in addition it also forms an internal water-mediated salt bridge with Glu32. These

interactions indicate the potential role of this residue in stabilizing the folding of the N-terminal

domain. However, the level of this effect may depend on the amino acid it is substituted with,

and its electrochemical properties. R261 has also been shown to destabilize the P-loop,

consequently decreasing ATP binding (Ban et al., 1999).

Page 97: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

80

Figure 2.2 A Ribbon diagram of the approximate position of human MLH1 R265 on the

crystal structure of E. coli MutL.

R265, which is equivalent to R261 in Escherichia coli was mapped onto the crystal structure of

the N-terminus of MutL (PDB#:1B63) using sequence homology. B. A more magnified view of

the amino acid R265 (E.coli R261): Indicates its likely involvement in stabilizing the folding and

the structure of the protein. C. R261 also forms an internal water-mediated salt bridge with

Glu32.

Page 98: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

81

Figure 2.3 Profile of time dependent expression of wild type MLH1 in HCT116 cells.

HCT116 cells were transiently transfected with 3 µg of pcDNA3.1+ expressing wt MLH1 and

harvested at the designated time points. Total extracts were immunoblotted with either anti-

MLH1 (upper panel) or anti-PMS2 (lower panel). An equal amount of the mismatch repair

proficient HEK293 total cell lysate was also immunoblotted as a positive control. Results from

untransfected (U) HCT116 cells indicate the absence of endogenous MLH1 expression.

Page 99: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

82

2.4.2 HCT116 cells are defective in MLH1

The colorectal cancer cell line, HCT116, was chosen as a model system for this study, as it is

deficient for endogenous MLH1 due to a hemizygous nonsense mutation in codon 252 [414].

The mutational status of these cells was confirmed by extracting RNA from the HCT116 cells,

reverse transcribing it and sequencing the resultant cDNA. We then confirmed the absence of

endogenous MLH1 at the protein level by performing western blotting assays as described

below. This indicated that the mutated endogenous MLH1 is rapidly degraded and therefore not

detectable by this assay, which is in line with previously published observations (Figure 2.3)

[390, 405].We also confirmed that PMS2, the cognate partner for MLH1 was present in these

cells in extremely low, almost undetectable levels, as it is unstable when not heterodimerized

with MLH1. We then transiently transfected in MLH1-pcDNA 3.1 vector using the

Lipofectamine 2000 reagent and assayed for its expression at various time points ranging from

12 hours to 7 days. This time course experiment indicated that we were able to transiently

express wild-type MLH1 from 12 hrs to 96 hrs post-transfection (Figure 2.3). However,

transfection of ectopic MLH1 restored endogenous levels of PMS2. This is consistent with

previous studies that indicate that PMS2 is unstable in the absence of MLH1 [260, 387, 390,

404].

2.4.3 R265C and K618A decrease expression of MLH1

Results from both tagged and non-tagged expression vectors indicate that all variant constructs

are capable of producing variant MLH1 protein of the expected size (~85kDa and ~ 90kDa

respectively). The R265C and K618A variants however, appear to either produce less protein or

Page 100: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

83

produce protein that is less stable than the wild-type MLH1 construct (Figure 2.4). On the other

hand, the expression profile for the variants I19F and L749Q were comparable to the wild-type

protein (Figure 2.4 and 2.5). No significant differences in β-galactosidase transfection efficiency

were seen between the wild-type and variant constructs. Representative data for K618A is

depicted in Fig. 2C. (Figure 2.5)

2.4.4 Altered expression is due to decreased stability of the protein

Differences in expression could occur due to a decrease in the levels of transcription and/or

translation, or due to an inherent decrease in the stability of the transcript or the protein. To

address whether the stability of MLH1 is altered by the substitutions R265C and K618A, we

employed pulse-chase assays to examine the rate of protein decay. Results indicate that R265C

and K618A have a half-life of ~ 5.5-6.5 and ~3-4 hours respectively. These variants are thus

significantly less stable than wild-type MLH1, which has a half-life about 10-10.5 hours (Figure

2.6). To verify these observations using an independent method; we treated transiently

transfected cells with cycloheximide, a global inhibitor of de novo protein synthesis, and

examined the stability of wild-type and variant MLH1 over time. Our experiments indicate that

the half-life of R265C and K618A MLH1 is significantly decreased compared to the wild-type

protein (Figure 2.7), thus corroborating our pulse chase results. To rule out technical influences

the variant L749Q, which shows comparable expression to wild-type MLH1, was included in the

cycloheximide assay. Our findings illustrate that the stability of the L749Q variant is comparable

to that of the wild-type protein (Figure 2.7). This finding validates our pulse chase results, and

demonstrates that our observations regarding decreased stability are due to the substitutions

introduced at codons 265 and 618 of the MLH1 gene.

Page 101: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

84

A.

B.

Figure 2.4 Expression of wild-type and variant MLH1 in HCT116 cells.

A. HCT116 cells were transiently transfected with wt or I19F MLH1 and harvested 24 h, 48h

and 72 h post transfection. The whole cell lysates (25µg) were immunoblotted for anti-MLH1

(upper panel) and anti-β-actin (lower panel). B. Cells were transfected with 3µg (A) or 5µg of wt

or R265C MLH1. U; Untransfected HCT116 cells, EV: empty vector. Results are representative

of at least 3 independent experiments.

Page 102: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

85

A.

B.

Figure 2.5 Expression profile of the MLH1 variants K618A & L749Q.

A. HCT116 cells were transiently transfected with 3µg of Myc-tagged wt, K618A or L749Q

MLH1 and harvested at 24, 48 and 72 hrs. Cell lysates were probed with antibodies for anti-Myc

(upper panel) and anti- -Actin (lower panel). Results indicate decreased expression of MLH1

K618A at all time points when compared to wt, while L749Q consistently showed comparable

expression to the wt vector. B. Transfection efficiency of K618A as assessed by β-galactosidase

activity: HCT116 cells were co-transfected with 0.15µg of pCMV β-galactosidase in addition to

the MLH1 constructs and β-galactosidase activity was measured in cell lysates. β-galactosidase

enzyme assays indicate that the K618A construct has a comparable, if not slightly higher,

transfection efficiency compared to that of the wt construct (comparable transfection efficiencies

were seen with the other variants). Data representative of at least 3 independent experiments and

error bars represent the 95% confidence intervals.

Page 103: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

86

Figure 2.6 Pulse chase assays to determine half-life of wild-type and variant protein.

HCT116 cells were transiently transfected with either wt, R265C or K618A MLH1 cDNA. The

amount of radiolabelled MLH1 pulled down at each time point was analyzed by densitometry,

normalized to -Actin, and plotted against time. The line of best fit for R265C/K618A MLH1

variants are depicted. As a reference, the line of best fit that corresponds to the half-life of wt

MLH1 is depicted on both graphs. Results indicate that the half-life of the variants R265C and

K618A is drastically altered (half-life of ~5.5 and ~3.5 hours respectively) compared to that of

the wt MLH1 protein (half-life of ~10 hours). Each experiment was performed a minimum of 3

times and representative results are shown.

Page 104: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

87

Figure 2.7 Stability of MLH1 variants as assessed by cycloheximide treatments.

HCT116 cells were transfected with either wt MLH1 or R265C, K618A, L749Q variant

constructs. Cells were then treated with cycloheximide (CXH) [+] or vehicle [-], 24 hours post-

transfection and lysed at indicated time points. MLH1 was detected by western blotting and β-

actin was probed for as a loading control. R265C and K618A MLH1 shows a significantly

reduced half-life (B, C), compared to wt MLH1 (A). L749Q however shows minimal effect on

stability (D), compared to the wt protein. 3 independent experiments carried out and

representative results are depicted.

Page 105: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

88

2.4.5 L749Q impairs ability of MLH1 to heterodimerize with PMS2

Interestingly, in our transfection experiments we also observed decreased levels of endogenous

PMS2 expressed when HCT116 cells were transfected with L749Q (Figure 2.8). We speculated

that this may be due to the decreased ability of MLH1 to bind to PMS2, due to the L749Q

missense substitution. Results from coimmunoprecipitation assays indicated that the amount of

PMS2 pulled down when transfected with L749Q is strikingly low compared to the wild-type

MLH1 protein (Figure 2.8). This indicates that this variant is unable to heterodimerize with

PMS2 as efficiently as wild-type MLH1. R265C and K618A variants, on the other hand, did not

appear to perturb the ability of MLH1 to heterodimerize with PMS2 (Figure 2.8). Taken together

these results indicate that the variant L749Q perturbs the ability of MLH1 to heterodimerize with

its partner PMS2, abrogates the formation of MutLα, despite having no observable effect on

expression.

Page 106: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

89

Figure 2.8 Ability of wild-type and variant MLH1 to interact with endogenous PMS2.

A. Levels of endogenous PMS2 expressed upon ectopic expression of MLH1 L749Q: HCT116

cells were transiently transfected with Myc-tagged wt or the L749Q MLH1 variant, and

harvested 24, 48 and 72 hrs post-transfection. Cell lysates were probed with anti-Myc (upper

panel) and anti-PMS2 antibodies (middle panel). -Actin was probed for as a loading control

(lower panel). Results indicate that as expected, endogenous PMS2 is stabilized in the presence

of wt MLH1. However, there is a considerable decrease in the levels of endogenous PMS2 levels

upon ectopic expression of the L749Q variant. B & C. Interaction of MLH1 variants with

endogenous PMS2 by coimmunoprecipitation experiments: HCT116 cells were transiently

transfected with 3µg of wt or variant MLH1 and harvested 48 hours post-transfection. Total

MLH1 was immnoprecipitated from 1000µg of whole cell lysate (WCL) using the MLH1

antibody. Levels of endogenous PMS2 and ectopic MLH1 pulled down in each sample were

probed for by western blotting. The upper and lower panels show PMS2 and MLH1 profiles

respectively, on ectopic expression of the respective MLH1 constructs. In the right panel, equal

amounts of WCL are loaded, to establish levels of protein. Expression of L749Q reveals

decreased PMS2 pulled down as an indicator of its disrupted ability to heterodimerize with

PMS2, albeit high levels of MLH1 expression (B). In contrast, the variants R265C and K618A

did not show disrupted ability heterodimerize with MLH1. As expected R265C and K618A

show decreased expression of MLH1 in the WCL samples when 25µg cell lysate are loaded.

However, there appeared to be no observable differences in the total level of MLH1

immunoprecipitated from 1000μg of cell lysates when transfected with wt, R265C or K618A,

likely due to the detection level of the antibody being saturated (C). Data representative of at

least 3 independent experiments.

Page 107: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

90

2.4.6 HCT116 cells express inducible wild-type and variant MLH1

A potential limitation of carrying out transient transfection experiments is that we cannot control

for the effects of variable transfection efficiency. Furthermore, transient transfection experiments

cannot be used to carry out treatments and measure phenotypic differences over longer periods of

time. Thus, we opted to generate stable inducible cell lines to assess the effects of these variants

on the functionality of MLH1. Inducible cell lines were chosen to minimize the variability that

could be introduced by the cells adapting to the presence of MLH1, as would be the case if an

uninducible stable cell line was generated. The major benefit of this model system therefore

would be that it would allow us to regulate the expression of MLH1. We generated four different

cell lines, EV, wtMLH1, I19F MLH1 and K618A MLH1. The empty vector cell line was

generated to help determine the contribution of other doxycycline responsive genes to the

phenotype measured. The I19F variant was chosen as the molecular mechanism by which this

substitution impacted MLH1 and MMR function was still unclear. In our previous assays it

neither affected MLH1 expression nor heterodimerization. Interestingly, when mapped to the

crystal structure of MLH1 it was located on the exterior surface of MLH1, and its location hinted

that it might be involved in interacting with other proteins. The K618A variant was chosen as it

showed altered protein stability, and as previous studies had indicated that DNA damage

signaling in response to an alkylating agent required full expression of MLH1 [311]. Clones that

show minimal levels of expression in the absence of treatment and maximal level of expression

upon induction were chosen and propagated. However, it was challenging to generate such

clones, especially those that successfully showed differential expression of wtMLH1. We able to

generate two such clones for the wtMLH1 cell line (out of about 90 clones screened), two for

K618A (out of about 30 clones) and three I19F (out of about 30 clones) (Figure 2.9, 2.10). Of

Page 108: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

91

these we chose the clone that showed maximal induction of wtMLH1 (clone 25); and variants

that showed comparable MLH1 expression to that of the wild-type clone (I19F-clone 20,

K618A-clone18). To further characterize these cell lines we induced cells with varying doses

(0.01-1μg/ml) of doxycycline to determine the lowest concentration of drug that yields robust

expression of wild-type and variant MLH1(Figure 2.11). All cell lines showed equivalent

induction at all concentrations. Thus, the lowest concentration tested, 0.01μg/ml was used in

subsequent experiments unless otherwise noted. We also carried out a time course of MLH1

induction in these cell lines, which showed a gradual increase in expression over time as

expected (Figure 2.11)

Page 109: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

92

A.

B.

Figure 2.9 Screening of clones.

Resistant clones of the Tet-repressor expressing cells or wild-type MLH1 expressing cells were

screened for by western blotting. Clones that showed the maximal induction of protein and the

least leakiness (assessed by minimal/negligent expression in the absence of Dox) were expanded

for subsequent experiments. [+] induced with Dox (doxycycline), [-] treated with vehicle, water.

Antibodies against the Myc-tag of MLH1 was used in these experiments.

Page 110: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

93

A.

IB: Actin

B.

Figure 2.10 Generation of the I19F MLH1 stable inducible cell line.

A. Stable clones generated for the I19F variant. [+] induced with Dox (doxycycline), [-] treated

with vehicle, water. B. Ribbon diagram of the approximate position of human MLH1 I19 on the

crystal structure of E. coli MutL: I19, which is equivalent to I14 in Escherichia coli was mapped

onto the crystal structure of the N-terminus of MutL (PDB#:1B63) using sequence homology.

Page 111: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

94

Figure 2.11 Characterization of the K618A stable inducible cell line.

A. The doxycycline concentration course for K618 and wild-type MLH1. Indicates that all

concentrations tested efficiently induced the expression of MLH1. B. Time course experiment

demonstrating induction of K618A MLH1 cell line. Similar profiles were seen with all stable

inducible cell lines with maximal induction evident as 24 hours post induction. U: Uninduced.

Page 112: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

95

2.4.7 Stable inducible cell lines and cell survival

When cell survival was assessed in the wtMLH1 stable inducible cell line in response to a range

of doses of temozolomide we observed no difference between induced and uninduced cells.

Similarly when the ratio of induced to uninduced cells expressing wild-type MLH1 was

compared to this ratio in the EV cell line no statistically significant differences were seen (Table

2.3). To determine if hampered expression was responsible for this lack of effect, we carried out

a western blot to confirm MLH1 expression in a subset of these conditions. Cells for western

blotting were plated and treated at the same time as the cell survival experiments and results

demonstrated unimpeded expression of MLH1 (Figure 2.12). Furthermore, these experiments

confirmed that the addition of temozolomide and O6-benzylguanine did not interfere with the

induction of MLH1 by doxycycline.

Page 113: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

96

Table 2.3 Summary of cell survival in response to temozolomide

Cell line

TMZ

dose

Relative Cell

# (I/U)

Std

Dev 95% CI P-value

EV 0 0.95 0.40 0.50 - 1.39

125 1.21 0.33 0.84 - 1.59

250 1.20 0.23 0.94 - 1.46

500 1.72 1.13 0.44 - 3.01

wt

MLH1 0 0.88 0.29 0.54 - 1.21 0.635

125 1.65 0.46 1.13 - 2.17 0.273

250 2.37 1.15 1.06 - 3.67 0.186

500 1.43 0.32 1.07 - 1.79 0.755

Data representative of 3 independent experiments. TMZ: Temozolomide, I: induced, U:

Uninduced, CI: confidence interval

Figure 2.12 Western blot to confirm levels of MLH1 in cell survival experiments.

TMZ: Temozolomide. BG: O6- Benzylguanine, Dox, Doxycycline.

Page 114: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

97

2.4.8 Stable inducible cell lines and the G2/M checkpoint

Previous studies have illustrated that MLH1 is important for the G2/M checkpoint arrest. In these

experiments we used the HCT116+Chr3 cell line as our positive control. This cell line contains

an extra copy of the entire chromosome 3 and as a consequence includes an extra copy of MLH1.

Thus, these cells express MLH1 and MMR assays have demonstrated that this cell line was able

to restore MMR proficiency when compared to MMR deficient HCT116 cell line [405].

Previous studies have demonstrated that this arrest is evident only when the phases of these cells

are synchronized prior to treatment with an alkylating agent [306]. Thus, in accordance with

these studies the cells were synchronized with low serum media (0.5% FBS) and then treated

with 100 or 500 µM temozolomide. At both these doses, 3 and 4 days after treatments the

HCT116+Chr3 cell line arrested in G2/M phase, while the parental HCT116 cell line did not do

so. However, the stable inducible cell lines expressing wt or variant MLH1 did not arrest 3 or 4

days after treatment (representative results depicted in Figure 2.13). Similar results were

observed in experiments carried out with varying concentrations of TMZ (100-500 µm).

Page 115: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

98

A. EV - Untreated

B. EV - TMZ Treated

C. HCT116+Chr3 - Untreated

D. HCT116+Chr 3 - TMZ Treated

Page 116: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

99

E. WT - Untreated

F. WT - TMZ Treated

G. I19F - Untreated

H. I19F - TMZ Treated

Page 117: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

100

I. K618A - Untreated

J. K618A - TMZ Treated

Figure 2.13 Representative cell cycle profiles of the stable inducible and HCT116+Chr3 cell

lines in response to treatment with temozolomide.

Cells were treated with 100 µM temozolomide, untreated cells were treated with an equal

concentration of vehicle, DMSO and the profile of the cells were assed 3 days later. The first

peak shows the relative proportion of cells in G1 phase expressing 2n DNA content. The second

peak shows the cells in G2 and M phases of the cell cycle expressing 4n DNA content. The red

arrow highlights the sample that displays a G2/M arrest profile. Representative results are

depicted here.

Page 118: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

101

2.4.9 Stable inducible expression of MLH1 & the microsatellite instability

phenotype of the HCT116 cell line

Given that the role of MLH1 in response to DNA damage is not as well defined, we wanted to

determine if stable inducible expression of MLH1 corrected the MSI-H phenotype of the

HCT116 cell line. The MSI phenotype is a surrogate marker of MMR activity and previous

studies have used similar approaches to determine the post-replicative error repair activity of

MMR proteins in cells [311]. However, when we assessed the stability of the BAT25 and

BAT26 markers, no change in instability was observed in the induced cells compared to

uninduced cells both at passage 10 and passage 30 (Table 2.4). Induction of MLH1 should

restore MMR activity, which should in turn restore stability at the BAT25/26 markers. Given that

the outcome of these experiments hinged on the continued expression of MLH1, we harvested

protein from a subset of cells at passage 20 and passage 30. Western blotting indicated that the

protein was continually expressed (Figure 2.14) and was therefore not responsible for this lack of

effect. However, the level of protein that was expressed by the cells appear to be lower at

passage 30 (at the conclusion of experiment) compared to the levels at passage 20.

Page 119: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

102

Table 2.4 Summary of MSI experiments

Cell line

Passage 0 Passage 10 Passage 30

Uninduced Induced

(Dox) Uninduced

Induced

(Dox)

wt MLH1 MSI-H MSI-H No change MSI-H No change

I19F MLH1 MSI-H MSI-H No change MSI-H No change

K618A MLH1 MSI-H MSI-H No change MSI-H No change

Grown independently for 30 generations either in the presence (induced) or absence (uninduced)

of Doxycycline (Dox). Cells were harvested at passage 0 (onset of experiment), 10, 30.

Page 120: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

103

Figure 2.14 Expression of MLH1 and PMS2 in wild-type MLH1 cells stable inducible cells.

Cell lysates were harvested after the EV and WT stable inducible cell lines were grown for 20 or

30 passages in the presence (I) or absence (U) of doxycycline. The upper panel shows MLH1

expression, the middle panel shows PMS2 expression and the lower panel shows the β-Actin

expression levels in these cells. HCT116+Chr 3 was included as a control to assess relative

expression levels.

Page 121: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

104

2.5 Discussion

The overall aim of this study was to employ a series of biochemical assays to elucidate if

missense variants in MLH1 are able to significantly alter protein expression/stability,

heterodimer formation and/or its function in signaling DNA damage. Our expression assays

illustrated that the variants R265C and K618A altered levels of MLH1 expression, leading us to

postulate that this might be due to decreased protein stability. Although, it has been previously

suggested that missense variants may decrease the stability of the MLH1 protein, to our

knowledge, no attempts have been made to date to assess the stability of the variant proteins

[391]. As several mechanisms may account for differences in expression, we chose to investigate

the stability of these variant proteins using both pulse-chase assays and cycloheximide

experiments. Both assays indicate that compared to wild-type MLH1, both R265C and K618A

missense substitutions decrease the half-life of the protein. It is important to note that we cannot

expect to observe identical half-lives for the respective proteins, given the technical differences

between the two protocols. The pathway responsible for the degradation of MLH1 is currently

unknown; therefore it is difficult to explain the mechanism responsible for this observed

decrease in stability. However, we speculate that missense substitutions may affect the tertiary

structure of the protein, which may lead to defective folding and the consequent degradation of

MLH1. The location of the residue on E. coli MutL and its putative interactions hint that such

might be the case for R265C.

In addition to the above strategies, we also used SIFT and PolyPhen to determine if these

programs can be used to predict the pathogenic nature of these variants. All three variants were

predicted to be deleterious, indicating that in the clinical setting these computational programs

are likely to be helpful in classifying the risk associated with novel variants. A recent study by

Page 122: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

105

Chan et al., demonstrated that the predictive value of these programs can be further increased by

using a combination of computational methods [386]. However, the two programs provided

discordant predictions with regard to K618A. Thus, in instances such as this where discordant

predictions are made, it remains important that missense alterations are assessed using a

combination of in-vitro/in-vivo strategies until improved models of prediction are available.

Several attempts have been made to assess the pathogenicity and/or biological significance of the

R265C and K618A variants. In this case R265C was identified in a proband that showed

deficient MLH1 expression by IHC, while the K618A variant was identified in a proband that

showed intact expression by IHC (details for IHC data included in Chapter 3). However, R265C

and K618A have been identified in multiple familes both in our clinic as well as others, and the

IHC findings from these families are not always identical. Quantitative MMR assays carried out

with R265C in Sacchromyces cerevisiae indicate that this variant retained its ability to repair

defects, albeit at lower proficiency than wild-type and „bona fide‟ mutants of MLH1[401]. These

observations lead the authors of this study to refer to this class of variants as „efficiency

polymorphisms‟. Interestingly, a study published less than a year later evaluated R265C using an

in vitro MMR assay and found it to be competent in post-replicative repair [385]. A recent study

employed conversion analysis technology to assess this variant. Conversion analysis is able to

expose masked mutations by converting the human chromosome complement to a haploid state

by fusing it with recipient E2 mouse cell line, after which these hybrid cell lines are used to

assay RNA expression. This study demonstrated that R265C leads to the skipping of exons 9 and

10, as well as a decrease in the levels of mRNA [234]. However, evidence for the skipping of

MLH1 exons 9 and 10 alone does not necessarily delineate the status of this variant as a disease

causing mutation; as previous studies have reported the existence of low levels of alternatively

spliced transcripts of MLH1 that lack these exons [415]. Therefore, it is likely that the

Page 123: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

106

detrimental effects of exon skipping depend on the level of the transcript that is deleted. It is also

possible that this and other variants contribute to HNPCC susceptibility by several mechanisms

including defective splicing and altered protein stability. Taken together these studies imply the

potentially pathogenic nature of this variant, and the associated risk for individuals carrying the

R265C variant. Our results provide further insight into the molecular basis of the pathogenicity

of R265C, by demonstrating that in addition to causing splicing defects, this variant may also

decrease the efficiency of MMR by destabilizing MLH1.

The K618A substitution replaces a charged amino acid with a neutral one. This is important as

K618 occurs alongside four charged amino acids that are well conserved in mammals, as well as

in Drosophila melanogaster [402]. Fearnhead et al., have proposed a link between the K618A

variant and an enhanced risk of multiple adenomas [402]. Additionally, in a recent study carried

out in the ovarian cancer cell line A2780, K618A showed normal levels of expression, but was

unable to reverse the mutator phenotype [400]. This variant was also unable to reverse tolerance

to methylating agents, a characteristic feature indicative of defective MMR. However, a study

carried out in the human embryonic kidney cell line 293T transfected with K618A demonstrated

that neither the expression of MLH1 nor the efficiency of repair activity as assessed by an in

vitro MMR assay was altered [383]. Furthermore, when Guerrette et al. examined the ability of

variants expressed in vitro to interact with PMS2 in a GST fusion protein interaction assay, they

found that K618A reduced the binding efficiency to PMS2 by > 85% [388]. In contrast to this,

we observed no effect of K618A on the ability of MLH1 to bind to PMS2 using

coimmunoprecipitation assays. Our observations are supported by the finding that in mlh1-/-

;

pms2-/-

murine embryonic fibroblasts, K618A did not display compromised ability to stabilize

PMS2 [404]. Moreover, results from yeast-two hybrid assays demonstrated that K618A was only

partially defective in its ability to interact with the partner proteins of MLH1 [403]. Although we

Page 124: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

107

cannot exclude the role of this variant in compromising binding and heterodimerization, it is

plausible that the differences in experimental conditions between the GST pull down assays and

coimmunoprecipitation assays may contribute to this discrepancy. Thus, based on these

functional assays alone it is difficult to determine if K618A is truly a pathogenic mutation or a

rare functional polymorphism. Our observation that K618A decreases the stability of the protein,

therefore offers an additional piece of evidence regarding the role of this variant in disease

susceptibility.

The discrepancies observed between these various analyses could be due in part to the diverse

experimental protocols, the specificity of cell types, and the sensitivity of the methods. We chose

to carry out our assay in a human colorectal cancer cell line to minimize such effects of cell type

or tissue specificity, and to enhance the relevance of our findings. In addition, another issue

which arises in the interpretation of findings from mismatch repair assays is the difficulty in

defining the threshold below which repair activity is abnormal or deficient. This leads to a

situation where researchers have to set up arbitrary cut-off values for normal MMR activity,

which influences the verdict of the role of these missense variants in compromising repair. It can

be postulated that irrespective of the functionality of the protein, decreased stability likely leads

to a situation where lower levels of protein are present in the cell. Although the levels of protein

essential for normal cellular function and the threshold below which cancer susceptibility occurs

has not been established; it is likely that decreased levels compromise to some degree function(s)

carried out by these protein complexes. For MLH1, possible functions affected in this manner are

not limited to the repair of mismatches, but may also extend to other functions such as DNA

damage signaling and recombination. It is also possible that varying threshold levels of MLH1

exist for the different functions of the MMR system, even in the same cells or tissues. The

observation that less MLH1 expression is required for the error-repair activity compared to the

Page 125: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

108

DNA damage response, might explain how these variants may still increase an individual‟s

susceptibility to HNPCC, without necessarily decreasing MMR activity. Thus, by establishing

that both R265C and K618A decrease the stability of the MLH1 significantly, our results now

demonstrate an independent mechanism by which the pathogenicity of these variants can be

established.

It is prerequisite that MLH1 heterodimerize with PMS2, to form the MutLα complex, prior to

carrying out any of its function described to date. Therefore, any variant that perturbs this

interaction could potentially compromise the efficiency of diverse functions carried out by this

complex. We observed that the variant L749Q disrupts binding to PMS2, despite proficient

MLH1 expression. This variant is located just outside the PMS2 interaction domain, but within

the C-terminal homology (CTH) domain of MLH1, which is exclusive to eukaryotic MutL

homologs [345, 387]. A study by Mohd et al., showed that a truncating mutation at the same

codon (MLH1 L749X) was capable of interacting with, but incapable of stabilizing PMS2 [404].

The results of this study lead the authors to conclude that the CTH domain plays a role in

stabilizing the MutLα complex, leading to the consequent stabilization of PMS2. Hence, it is

possible that a similar mechanism may underlie our observations regarding the L749Q variant.

Previous studies using a cell line with inducible MLH1 has shown that while post-replicative

error repair can occur even when as little as 10% of MLH1 is expressed, DNA damage signaling

in response to MNNG requires full expression of MLH1[311]. This indicates the possibility that

MLH1 variants that show differences in stability may have differential levels of

resistance/tolerance to chemotherapeutic agents, which can be exploited to study the role of

MLH1 in these processes. We generated stable inducible cell lines of wild-type and variant

MLH1 in order to minimize the confounding effects that could occur due to variable, transient

Page 126: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

109

expression. Cells deficient in MMR are tolerant to alkylating drugs such as temozolomide and

treating MMR proficient cells with increasing concentrations of temozolomide was shown to

lead to decreased cell survival [314, 416]. Despite continued expression of MLH1, we did not

see altered cell survival and/or G2/M cell cycle arrest in the stable inducible cell lines expressing

wild-type or variant MLH1. The positive control HCT116 + Chr3 has been shown to display

MLH1 expression and correct the MSI-H phenotype of these cells [405]. In contrast to our stable

inducible cells, treatment with temozolomide induced G2/M arrest in the HCT116+Chr3 cells,

which is in agreement with previously published studies. In our MSI experiments we did not

observe a change in the level of stability at the BAT25 or BAT26 loci, despite sustained MLH1

expression. However, this result should be interpreted cautiously, as given the nature of these

experiments (the requirement to serially passage cells in the presence of doxycycline) we did not

have an appropriate positive control. Thus, it appears that although the stable inducible cell lines

generated express MLH1, the functionality of this protein is limited. Previous attempts to

generate HCT116 with stable expression of MLH1 led to a growth inhibitory effect and the

inability to restore MMR proficiency despite the detectable expression of the MLH1 protein

(unpublished results, personal communication Dr. Bert Vogelstein). Furthermore, in a previously

published study using a stable inducible cell line in the HEK293T background, expression of

MLH1 resulted in G2/M arrest [311]. However, it appears that even in this cell line the

sensitivity to damaging agents was not evident in clones that showed overexpression of MLH1

compared to PMS2; and only in cases where PMS2 expression was comparable to MMR

proficient cell lines (in this case the parental cell line 293T) did they observe MNNG sensitivity

(personal communication Dr. P. Cejka). Based on these findings the most likely reason for this

lack of response is that the overexpression of MLH1 negatively affects the cell and possibly

disrupts the function of the MMR system. A similar phenotype has been described in

Page 127: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

110

Saccharomyces cerevisiae, where the overexpression of MLH1 was shown to lead to the mutator

phenotype and defective MMR [417]. A possible mechanism for this is that the overexpresed

MLH1 is not able to heterodimerize with PMS2 efficiently and the remaining excess partner-less

MLH1 disrupts MMR activity in this in vitro system. The low level of PMS2 stabilized by

MLH1 in these cell lines is evident in Figure 2.14. A possible explanation for MLH1‟s inability

to heterodimerize with PMS2 could be that the Myc-tag interferes with correct protein folding.

Therefore, these studies indicate that the level of MLH1 is carefully regulated in cells. Thus, a

biologically meaningful increase or decrease in MLH1 expression impairs the functions carried

out by the MMR system.

In summary, we observed that the two naturally occurring variants R265C and K618A decreased

the stability of MLH1. In addition a third variant generated by us, L749Q, affected MLH1‟s

ability to heterodimerize with PMS2. Although no single test or assay can definitively indicate

the pathogenicity of a variant, combined data from a series of assays with regard to a specific

variant or to comparable variants that occur in close proximity will likely help elucidate the

pathogenicity associated with unclassified variants identified in HNPCC patients.

Acknowledgements

We would like to thank Dr. Yosr Haffani for her assistance with mapping the variants on the

crystal structures. We would also like to thank Dr. Ken Lau, Annie Bang, Kelly Fenwick and

Ted Chappell for their technical assistance.

Page 128: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

111

CHAPTER 3

A novel method of determining the effect of MLH1 unclassified genetic

variants on differential allelic expression

Sheron Perera1, 2, 3

, Brian Li2, Soultana Tsitsikotas

3, Aaron Pollett

1, 3, Kara Semotiuk

4, and

Bharati Bapat1, 2, 3

1 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON,

Canada; 2

Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, ON, Canada;

3 Department of Pathology and Laboratory Medicine, Mt. Sinai Hospital, Toronto, ON, Canada;

4 Dr. Zane Cohen Digestive Diseases Clinical Research Centre, Mt. Sinai Hospital, Toronto, ON,

Canada

The work in this chapter was primarily contributed by Sheron Perera. BL, an undergraduate 4th

year project student worked with SP and helped with DNA and RNA extractions. ST, a

technologist extracted RNA from a subset of patient lymphocyte samples. AP, a pathologist

confirmed IHC staining patterns and KS, a genetic counselor provided access to family history

and clinical information of the probands.

The data presented in this chapter is currently in press: Perera, S., Li, B., Tsitsikotas, S., Pollett,

A., Semotiuk, K., and Bapat, B. A novel and rapid method of determining the effect of MLH1

unclassified genetic variants on differential allelic expression Journal of Molecular Diagnostics

(2010).

Page 129: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

112

3 Chapter 3 A novel and rapid method of determining the

effect of MLH1 unclassified genetic variants on differential

allelic expression

3.1 Summary

Germline mutations in mismatch repair genes predispose patients to Lynch Syndrome and the

majority of these mutations are seen in two key genes, MLH1 and MSH2. In particular, about a

third of the missense variants identified in MLH1 are of unknown clinical significance. Using the

PeakPicker program we have conducted a proof-of-principle study to investigate whether

missense variants in MLH1 leads to allelic imbalances. Lymphocyte RNA extracted from

patients harboring known MLH1 variants was used to quantify the ratio of variant to wild-type

transcript, while lymphocyte DNA was used to establish baseline allelic expression levels. Our

analysis indicates that the missense variants c.350C>T, c.793C>T, c.1852_1853AA>GC, as well

as the truncating variant c.1528C>T led to significantly unbalanced allelic expression. However,

the variants c.55A>T and c.2246T>C did not demonstrate an allelic imbalance. This illustrates a

novel and efficient method of investigating the pathogenicity of unclassified genetic variants

discovered in mismatch repair genes, as well as genes implicated in other inherited diseases. In

addition, the PeakPicker methodology has the potential to be applied in the diagnostic setting,

which in conjunction with results from other assays will help increase the accuracy and

efficiency of genetic testing of colorectal cancer, as well as other inherited diseases.

Page 130: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

113

3.2 Introduction

Lynch syndrome or hereditary nonpolyposis colorectal cancer (HNPCC; MIM 120435),

which accounts for about 2% of all colorectal cancer cases, is one of the most common cancer

predisposition syndromes. Those affected have an 80% lifetime risk of developing colorectal

cancer[233]. In addition, malignancies of the endometrium, skin, bladder, ovaries, kidney, and

small intestine are also associated with this syndrome.

Lynch syndrome is an autosomal dominant condition that is characterized by germline

mutations in mismatch repair (MMR) genes. The MMR system is a post-replicative DNA repair

system that recognizes and repairs base-base mismatches and insertion/deletion loops that occur

during DNA replication and escape the proofreading activity of the DNA polymerases. MMR

proteins have also been implicated in signaling DNA damage and apoptosis. Germline mutations

of the MMR genes MLH1 (MIM 120436), MSH2 (MIM 609309), PMS2 (MIM 600259), PMS1

(MIM 600258), and MSH6 (MIM 600678) have hitherto been implicated in Lynch syndrome.

However, about 90% of all known mutations are found in the two MMR genes MLH1 and

MSH2[397], which appear to play essential and non-redundant functions of the MMR system.

Tumours with defective MMR function are characterized by expansion/contraction of

the microsatellite regions, leading to microsatellite instability (MSI), a hallmark feature seen in

about 85% of Lynch syndrome tumours[123]. Individuals diagnosed at a younger than average

age and/or with familial clustering of colorectal or other Lynch syndrome associated cancers are

generally referred to predictive genetic testing programs. Here, MSI testing is carried out on

tumour DNA, which acts as a surrogate marker of defective MMR function. Typically,

immunohistochemistry (IHC) is also performed to assess if MMR protein expression is lost.

Page 131: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

114

Although MSI testing is a robust assay of mismatch repair activity, it is not a universal indicator

of pathogenicity as it is possible for mutations to affect other functions of the MMR pathway

[418]. Similarly, the loss of protein expression by IHC does not definitively link the unclassified

variant to the loss of protein expression. Additionally, some tumours show equivocal staining

patterns with intermediate levels of expression, which cannot be clearly distinguished by IHC.

Furthermore, methylation of the MLH1 leads to the MSI phenotype and the loss of MLH1

expression in about 15% of all sporadic colorectal cancers [121, 122]. Thus, to confirm the

presence of Lynch syndrome, the MMR genes of these individuals are screened for germline

mutations. Similar to the other MMR genes, diverse sequence variants, including nonsense,

missense, splice site, and frameshift mutations, have been reported throughout the coding region

of the MLH1 gene. Many of these mutations are catalogued in the LOVD and MMR Variant

Databases (http://chromium.liacs.nl/LOVD2/colon_cancer/home.php and

http://www.med.mun.ca/MMRvariants, last accessed December 16, 2009) [372] About 24% of

mutations detected in MLH1 are missense substitutions [372].The majority of these missense

variants are of unknown clinical significance. Given that the diagnosis of Lynch syndrome relies

primarily on the identification of germline defects in the MMR genes [234], detecting these

unclassified genetic variants creates much ambiguity in the clinical setting as the pathogenicity

of these variants cannot be readily ascertained.

Upon detection of missense variants several strategies are employed to characterize their

pathogenic significance. These include linkage analysis, association studies, computational

prediction tools and functional assays, each with specific strengths and limitations [234, 382-

385, 389, 390]. Linkage analyses is often the most straightforward way to determine the

relationship between the disease phenotype and the segregation of the variant, but is often not

feasible due to smaller family sizes or unavailability of samples from relevant family members.

Page 132: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

115

Computational programs, while helpful in classifying novel variants in the clinical setting, are

limited in their predictive value unless a panel of such programs are used[386]. However, at

present it is important to use a combination of in silico and experimental strategies until

improved models of prediction become available. We and other groups have attempted to devise

in vitro assays to characterize the pathogenic and functional significance of MLH1 variants. A

common approach is to create these alterations in vitro by site-directed mutagenesis, after which

these constructs are used to assay expression, MMR function or protein stability [383, 385, 389-

391]. However, this approach requires a considerable investment of time and effort and cannot

be routinely performed in a clinical diagnostic service.

Determining unbalanced allelic expression offers an alternative method of investigating the

pathogenic nature of a variant. Several studies in other genes have demonstrated that single

nucleotide substitutions are capable of altering the levels of mRNA transcripts [419-421].

Furthermore it has been proposed that such cis-acting functional variants can affect transcription,

messenger RNA (mRNA) processing and mRNA stability [420]. We have conducted a proof-of-

principle study to investigate whether missense variants in MLH1 lead to an allelic imbalance by

measuring levels of transcript present in lymphocytes of individuals carrying the variants

c.55A>T (p.I19F), c. 350C>T (p.T117M), c.793C>T (p.R265C), c.1528C>T (p.Q510X),

c.1852_1853AA>GC (p.K618A), and c.2246T>C (p.L749P), using the PeakPicker method.

3.3 Materials & Methods

3.3.1 Selection of Patients and Families

These probands were referred to the Molecular Genetics Laboratory as part of the Provincial

Cancer Genetics Program for the assessment of the possible diagnosis of Lynch syndrome.

Predictive genetic testing was offered to clinically affected and at risk subjects, with pre- and

Page 133: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

116

post-test genetic counseling as described previously[422]. In addition to complete follow-up

information, clinical and histopathological data were collected retrospectively

on all affected

patients. Informed consent was obtained from all subjects and all studies were performed

according to guidelines of the Ethics Committee of the University of Toronto.

3.3.2 Overview of diagnostic strategy and identification of MLH1 variants

The variants chosen for this study were identified in patients that were screened through the

Ontario Predictive Genetic Testing Program at Mt. Sinai Hospital, Toronto, Canada based on a

series of predetermined referral criteria. This program screens individuals presenting with either

a family history of colorectal cancer, and/or those who develop tumours at a young age of onset

(<50). Genetic screening is initiated by performing MSI testing and IHC for the MMR proteins

on tumour specimens when available. This is followed by germline mutation testing on

lymphocyte DNA. Initially, multiplex ligation-dependent probe amplification (MLPA) is

performed to detect large genomic deletions and duplications in the MMR genes. If such changes

are not detected, germline DNA is screened by sequencing the MLH1 and MSH2 genes, guided

by MSI/IHC results of patient tumours. Through this program we have identified many

established and novel germline mutations, as well as several unclassified variants.

3.3.3 Microsatellite instability and immunohistochemistry

MSI testing and IHC analysis for MMR proteins were performed as described previously.[410]

Matched normal and tumour DNA were assessed using a panel of five microsatellite markers as

recommended by the National Cancer Institute[232]. These include –mononucleotide BAT25

and BAT 26 and dinucleotide D2S123, D5S346 and D17S250 microsatellite markers. Each case

was designated as either microsatellite unstable (MSI-H; ≥30% markers unstable),

MSI-low

(MSI-L; < 30% markers unstable), or microsatellite stable (MSS; no unstable markers). IHC

Page 134: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

117

analysis of the respective MMR proteins was performed on formalin-fixed, paraffin-embedded

tissues.

3.3.4 DNA extraction and mutation detection

Blood samples were obtained from patients and lymphocytes were isolated using NH4Cl–Tris.

DNA was extracted from lymphocytes using the QIAGEN or saturated salt-out method as

described previously [408]. DNA from patients, whose tumours showed deficiency for a MMR

protein, was subjected to exon by exon sequencing of genomic DNA to screen for alterations in

MLH1 and MSH2 on an ABI 377 DNA Sequencer (Applied Biosystems, Foster City, CA).

Sequence information of the coding region was derived from RefSeq NM_000249.2 (MLH1),

NM_000251.1 (MSH2). PCR conditions and primer sequences are available upon request.

3.3.5 RNA extraction and cDNA synthesis

RNA extraction was performed using TRIzol according to the manufacturer‟s protocol

(Invitrogen Life Technologies, Burlington, ON). The RNA then treated with DNaseI for 30

minutes using the RNase-Free DNase set (Qiagen), which allows for digestion of DNase prior to

purification using the RNeasy MinElute kit (Qiagen). Reverse transcription PCR was performed

according to standard techniques as described before [408] and cDNA was generated in triplicate

from each RNA sample to minimize variation that may be introduced during the process of

reverse transcription.

3.3.6 PeakPicker Analysis

3.3.6.1 Primer design and PCR

Primers were designed to amplify the exonic regions flanking the variant of interest so as to

allow the same pair of primers to be used for the amplification of both the genomic (gDNA) and

complementary DNA (cDNA) to ensure unbiased amplification (Table 3.1). PCR products were

Page 135: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

118

then visualized on an 1.7% ethidium bromide/agarose to verify the presence of product. PCR

products were treated with 5 units of Shrimp Alkaline Phosphotase and 2 units of Exonuclease I

per 15µl of PCR product for 37°C for 1 hour to remove unincorporated dNTPs and excess

amplification primers. The enzymes were then deactivated by incubating at 75°C for 15 min.

3.3.6.2 Sequencing and PeakPicker Analysis

High sensitivity sequencing was carried out as described before [423], using 0.5 µL of Big Dye

Terminator v3.1 Cycle Sequencing Kit (Applied Biosystems) with 2 µL of PCR product , 1.75

µL of 5x sequencing buffer and 10pmol of sequencing primer in a 10 µL reaction using the

manufacturer‟s protocol. The products were separated on an Applied Biosystems 3130XL DNA

analyzer. Sequencing was carried out with both the forward and reverse primers whenever

possible, unless the variant was located too close to the start of the primer.

The PeakPicker software was developed by Ge et al., for quantitative allele ratio analysis [423,

424]. This software is publicly available at http://genomequebec.mcgill.ca/publications/pastinen/

(last accessed April 1, 2010). It relies on multiple alignments of sequence traces generated by

high sensitivity sequencing. The reference sequence (gDNA) is used to select the peaks that

represent the single nucleotide polymorphism (SNP) or variant, as well as control peaks. The

program then identifies SNP and control peaks in all sequences, and analyzes the peaks in

parallel. The allelic ratio of the gDNA samples are set to 1 as no allele specific expression is

expected. However, by concurrently analyzing the gDNA sample in triplicate, we were able to

increase the accuracy of this normalization step. A file with SNP allele ratios normalized to

gDNA peak heights is generated as the output of this program. Allelic ratios were calculated by

dividing the peak height of the variant allele by that of wild-type allele. Three independent PCR

and sequencing reactions were carried out from each gDNA and RNA (one reaction from each

Page 136: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

119

independent cDNA) sample and a two-tailed Student‟s t-test was used to assess statistically

significant differences in allelic expression.

3.3.7 Allele specific (Asymmetric) PCR

Primers were designed to amplify the exonic regions surrounding the variant and the c. 655A>G

polymorphism. Differential amplification of alleles was achieved by designing two unique

reverse primers that contained either of polymorphic bases at its 3‟ end. Given the stringent

requirement for complementarity at this location, this allowed us to preferentially amplify each

of the alleles. PCR was then carried out as described above using a common forward primer,

except that twice as much reverse primer was used as the forward primer to ensure successful

amplification of the product. PCR products were then purified with Shrimp Alkaline

Phosphotase and Exonuclease and sequenced using the Applied Biosystems 3130XL DNA

analyzer.

3.3.8 In silico analysis of potential effects on splicing

We used the computational programs NNSPLICE (http://www.fruitfly.org/seq_tools/splice.html,

last accessed April 13, 2010) and GeneScan (http://genes.mit.edu/GENSCAN.html, last accessed

April 13, 2010) to determine the predicted effect of the splice site mutation. We also used the

program ESEfinder (http://rulai.cshl.edu/cgi-bin/tools/ESE3/esefinder.cgi?process=home, last

accessed April 19, 2010), RescueESE (genes.mit.edu/burgelab/rescue-ese/,last accessed April 19,

2010 ) and PESX (http://cubweb.biology.columbia.edu/pesx/, last accessed April 19, 2010) to

assess if these variants created or abolished exonic splice enhancer (ESE) or exonic splice

silencer (ESS) sites.[425, 426] Default settings were used for all programs.

Page 137: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

120

Table 3.1 Primers used for PeakPicker analysis

Variant/Exon

Primers (5’-3’)

I19F/1

F: GGTTGAACATCTAGACGTTTCCT

R:TTCTCAATCATCTCTTTGATAGCA

T117M/4

F: ATATAT CTT TGG CCA GCA TAA GCC

R: CTG TAT GCA CAC TTT CCA TC

I219V/8

F: CAA GGA GAG ACA GTA GCT GA

R:CGACTAACAGCATTTCCAA

Q510X/13

F: TCTGATGTGGAAATGGTGGA

R: CCTCATGTCCCTGCTCATTA

CATGTCCCTGCTCATTAATTTCTTCCT

K618A/16

F:CAGCACCGCTCTTTGACC

R: TTTCCAAAGAGAAATAGTCTGCAA

L749P/19

F: CCAAACTCCTGGAAGTGGAC

R: TCCCACAGTGCATAAATAACCA

Primers surrounding the R265C variant could not be designed as it was located to the intron-

exon boundary, thus primers surround I219V was used. F: Forward primer, R: Reverse primer

Page 138: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

121

3.4 Results

3.4.1 The truncating variant c. 1528C>T leads to unbalanced allelic expression

In this study we determined the allelic expression of MLH1 in lymphocytes obtained

from individuals carrying missense variants c.55A>T, c.350C>T, c.793C>T,

c.1852_1853AA>GC, c.2246T>C and the truncating variant c.1528C>T in MLH1. Genomic

DNA (gDNA) was used as a reference to establish baseline levels of expression, and cDNA

generated from three independent reverse transcription reactions was used to measure mRNA

transcript levels. Identical primer pairs were used to PCR amplify the region surrounding the

variant in both gDNA and cDNA samples, followed by high sensitivity sequencing of the

amplicons (Figure 3.1). The sequence information was then analyzed using the PeakPicker

program. This program quantifies peak heights of the two polymorphic bases and uses this

information to compute their relative abundance in the sample. As a component of this step this

program compares the heights of adjacent peaks to mitigate any effects the efficiency of the

sequencing reaction might have on overall peak height. The ratio of two variant bases is set to 1

in the gDNA samples and the heights of the peaks are normalized to the values derived from

gDNA. In accordance with previously published studies, we set ≤ 0.5 as a threshold value below

which allelic ratios were considered abnormal [427, 428]. This ratio corresponds to a situation

where twice as much wild-type transcript is present compared to the transcript carrying the

variant base.

Case 4 was diagnosed with CRC at the age of 42. Exonic sequencing indicated that this

individual carries the nonsense mutation c.1528CT in exon 13 of MLH1. This mutation led to

an allelic ratio of 0.38 (p = 0.0023). Results generated from PeakPicker analysis, as well as

clinicopathological variables are summarized in Table 3.2 and Table 3.3 respectively.

Page 139: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

122

3.4.2 The missense variants c. 350C>T, C. 793C>T and c.1852_1853AA>GC

lead to unbalanced allelic expression

Case 2 presented for at-risk testing and is a carrier of the c. 350C>T variant, which occurs

in exon 4. Her father (the proband) was diagnosed with MSI-H, MLH1 deficient adenocarcinoma

of the colon and was found to carry c. 350C>T. However, his lymphocyte DNA and RNA was

not available for analysis. This variant led to an allelic ratio of 0.19 (P = 0.015) (Table 3.2). Case

2 was also heterozygous for the c.655A>G (p.I219V) polymorphism. We therefore reanalyzed

this case using a primer set that flanked this polymorphism to rule out the possibility that the

observed allele ratio was primer specific. When PeakPicker analysis was carried out using

primers that flanked this polymorphism in exon 8 of MLH1, the allelic ratio was found to be

0.07(P = 0.0004), which is in agreement with the results obtained using primers that flank the

variant in exon 4.

The variant c.793C>T, which occurs in exon 10 was detected in an individual who

developed CRC at age 55(case 3). Given the location of this substitution in the third nucleotide at

the beginning of the exon, it was not possible to use identical primers to amplify the region

surrounding this variant from both gDNA and cDNA. However, case 3 was heterozygous for the

c.655A>G polymorphism, which enabled us to use primers flanking this region to assay its

allelic ratio. Using allele, specific PCR we were able to show that c.655A occurs on the wild-

type allele (c.793C). PeakPicker analysis demonstrated that this variant was associated with an

allelic ratio 0.5 (P = 0.025).

The MLH1 variant c.1852_1853AA>GC was the only genetic alteration discovered in a

proband that developed small bowel cancer at the age of 34 years (Case 5A). This variant

represents a two nucleotide substitution in exon 16 of MLH1. PeakPicker analysis revealed that

the proband‟s allelic ratio was reduced to 0.05 (P = 0.0009) (Figure 3.1). This proband was also

Page 140: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

123

heterozygous for a common MLH1 polymorphism c.655A>G (where c.655A occurs on the wild-

type allele). Using primers that flank this polymorphic region the MLH1 allelic ratio was

determined to be 0.13 (P = 0.0004), confirming our previous finding. This proband‟s father (case

5B), who presented for at-risk testing, is also a carrier of c.1852_1853AA>GC variant. By

PeakPicker analysis case 5B‟s MLH1 allelic ratio was demonstrated to be 0.5 (P = 0.0135). In

keeping with the results from case 5A, this variant appears to be associated with an allelic

imbalance. However, as this individual was not heterozygous for the c.655A>G polymorphism

we were unable to repeat the analysis as with case 5A. The predicted effects of these alterations

on splicing are summarized in table 3.4.

3.4.3 The missense variants c. 55A>T and c. 2246T>C do not lead to

unbalanced allelic expression

By exonic sequencing we identified the missense variant c.55AT in exon 1 of an

individual who developed endometrial cancer at the age of 47 (case 1). Results from the

PeakPicker analysis demonstrated that the allelic expression of the variant allele was 0.86 (p =

0.564) (Table 3.2), which was not found to be statistically significant when assessed using a two-

tailed Student‟s t-test.

The c.2246TC variant was discovered in a proband who developed colorectal cancer at

the age of 24 (case 6). This variant occurs in exon 19 of MLH1 and led to an allelic ratio of 1.02

(p = 0.8253, Figure 3.2).

Page 141: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

124

Figure 3.1 Representative chromatograms generated by high sensitivity sequencing.

The left panels (A, C) indicate genomic DNA (gDNA) and the right panels (B, D) indicate

complementary DNA (cDNA). Case 5A demonstrates decreased levels of the variant (GC) allele

relative to the wild-type(AA) allele, while case 6 shows no change in the levels of the variant(C)

allele relative to the wild-type (T) allele. Arrows highlight the heterozygous peaks. Both

chromatograms shown were generated using the respective forward primer. (Blue: C, Green: A,

Red: T, Black: G)

Page 142: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

125

Table 3.2 Summary of allele ratios generated by PeakPicker

Case Sample

Av. variant/wt

peak heightA

95% CI

Allele

Ratio

P-

value

1 gDNA 1 (0.96-1.05) 0.85 0.5640

cDNA 0.86 (0.47-1.25)

2 gDNA 1.02 (0.82-1.23) 0.19 0.0155

cDNA 0.2 (0.17-0.22)

2# gDNA 1 NC 0.07 0.0004

cDNA 0.07 (0.04-0.11)

3 gDNA 1 (0.97-1.05) 0.5 0.0256

cDNA 0.5 (0.32-0.56)

4 gDNA 1 (0.97-1.03) 0.38 0.0023

cDNA 0.38 (0.31-0.46)

5A gDNA 1 (0.94-1.07) 0.05 0.0009

cDNA 0.05 (0.03-0.06)

5A# gDNA 1 (0.97-1.03) 0.13 0.0004

cDNA 0.13 (0.12-0.13)

5B gDNA 1 (0.93-1.08) 0.5 0.0135

cDNA 0.5 (0.39-0.61)

6 gDNA 1 (0.98-1.02) 1.02 0.8250

cDNA 1.02 (0.9-1.13)

A

Average height of variant peak/ height of wild-type peak

Allele ratios = Average height of variant peak/ height of wild-type peak in cDNA

Average height of variant peak/ height of wild-type peak in gDNA

P –values are calculated using a two-sided Student‟s t-test. P -values ≤ 0.05 are shown in bold.

All PCR reactions were carried out with primers that flank the region surrounding the variant,

unless indicated by a # sign, which denotes results obtained using primers that flank the

polymorphic region on Exon 8. CI: Confidence interval, WT: wild-type, NC: Not computable as

replicates generated identical values.

Page 143: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

126

Table 3.3 Summary of clinicopathological characteristics

Case Alteration

Predicted

effect on

protein

Clinical

diagnosis

MLH1

IHC

Status

Tumour

MSI

status

1 c.55A>T Ile19Phe

Endometrial

cancer at 47 ND ND

2 c.350C>T Thr117Met

At-risk

testing a N/A N/A

3 c.793C>T Arg265Cys CRC at 55 Deficient MSI-H

4 c.1528C>T Gln510Ter CRC at 42 Deficient MSI-H

5A c.1852_1853AA>GC Lys618Ala

Small bowel

cancer at 34 Intact MSI-H

5B c.1852_1853AA>GC Lys618Ala

At-risk

testing N/A N/A

6 c.2246T>C Leu749Pro CRC at 24 Intact MSI-H

a Case 2‟s father ( the proband) carried the same variant and presented with a MLH1 deficient,

MSI-H tumour but his DNA/RNA was not available for analysis. 5A and 5B are members of the

same kindred. IHC: immunohistochemistry; Intact: intact expression of MLH1 by IHC;

Deficient: MLH1 expression absent by IHC; MSI-H: Microsatellite instability-high; ND: Not

determined (No access to tumour sample); CRC: colorectal cancer; N/A: Not applicable

Page 144: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

127

Table 3.4 Predicted effects of missense variants on splicing

MLH1

Variant AI

Effect on

Splicing

Rescue

ESEa

PESXb

ESEfinder c

SF2 SC35 SRp55 SRp40

c.55A>T No NC NC - 3.07/ - 4.7/3.0 -

c.350C>T* Yes 4/2

1/0

ESE - - - 4.60/3.06

c.793C>T* Yes

dExon

skipping NC NC - - - 3.85/4.21

c.1852_1853

AA>GC* Yes 4/1

4/1

ESE

-

/2.38 - /2.86 - -

c.2256T>C No NC NC - 3.04/3.8 - /2.86 -

* These in silico predictions have been published in previous reports that assessed the impact of

unclassified variants on splicing [429, 430]

a The number of ESE motifs predicted by Rescue ESE for the wild-type sequence/ variant

sequence b

The number of ESE/ESS motifs predicted by PESX for the wild-type sequence/ variant

sequence c The score for ESE motifs recognized by 4 SR proteins as predicted by ESEFinder for wild-

type/variant d

This variant has been report to lead to the skipping of exon 10[430]

AI: Allelic imbalance, defined as having a allelic ratio ≤0.5 by PeakPicker analysis; NC: No

change predicted;

Page 145: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

128

3.5 Discussion

The discovery of a predisposing mutation in Lynch syndrome families and the concomitant

increase in surveillance has been shown to significantly reduce mortality [251, 431]. Therefore it

is of paramount importance to classify the pathogenicity associated with unclassified variants to

determine their role in cancer predisposition. PeakPicker analysis provides a relatively rapid and

robust method of differentiating potentially pathogenic variants that lead to unbalanced allelic

expression. Furthermore, no specialized tools or complex technical skills are required to perform

this analysis, thus this technique has the potential to be implemented in a diagnostic setting for

the routine assessment of unclassified genetic variants.

The c.55AT variant has been reported in two previous independent studies; however, no

functional studies have been carried out to date [398, 399]. Although PeakPicker analysis

demonstrated that this variant generated an allelic ratio that was less than 1, this value was not

found to be statistically significant. In this study considered an allelic ratio ≤ 0.5, combined with

statistical significance to be indicative of altered allelic expression/stability. A previous study

documented that the normal range of allelic variation in MLH1 is between 0.81 and 1.23[432],

which is in line with our observation that this variant likely does not lead to an allelic imbalance.

It is possible that some genetic variants may only mildly affect mRNA levels and thus lead to

intermediate allelic ratios (1 - 0.5). The accurate interpretation of these variants in the clinical

setting will require that the arbitrary cut off established in this study be considered in the light of

other available evidence (i.e. MSI/IHC information of the proband‟s tumour). In this case we did

not have supplementary data from MSI and IHC. However, we observed expression levels

comparable to that of wild-type MLH1 protein by in vitro expression assays (Figure 2.4). Thus,

combined these observations suggest that altered mRNA expression/stability is unlikely to be the

Page 146: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

129

mechanism by which this variant mediates its pathogenicity but highlights the utility of

PeakPicker analysis in cases where incomplete clinical information is present.

Based on phylogenetic, structural and functional evidence, as well as segregation data the

c.350 C>T variant is usually considered to be a deleterious mutation [430]. This variant has been

shown to affect the efficiency of MMR function by several studies [413, 433]. However by

conversion analysis technology this variant (detected in a proband from a separate family) did

not show decreased allelic expression [234]. Conversion analysis separates alleles by converting

the human chromosome complement to a haploid state by fusing it with recipient E2 mouse cell

line. The resulting hybrid cell lines contain only a subset of human chromosomes and are used to

assay cDNA sequence changes. While the benefit of this approach is that it allows the maternal

and paternal alleles to be analzysed independently, the hybrid nature of the cell may affect the

regulation and expression of mRNA. Thus, this discrepancy may have arisen due to the

differences between these two methodologies, another possibility is that other unique genetic

influences may have led to the distinct allelic profiles observed in the two individuals.

The c.793C>T variant have been reported to lead to reduced MMR efficiency in several

assays [401, 413, 433, 434]. Furthermore, in experiments in carries out in our laboratory we were

able to show that this variant decreased the stability of the MLH1 protein [389]. This variant has

also been associated with abnormal splicing, with two independent studies demonstrating that it

induced partial skipping of exon 10 (Table 3.4) [429, 430]. Given its proximity to the intron-

exon border this highlights the role of cis-acting elements in flanking sequences in regulating

splicing and defining exons. Interestingly, this variant also decreased the expression of the

variant allele when measured using conversion analysis [234]. Given the distinctions of the two

Page 147: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

130

assays, this finding provides evidence of the sensitivity and accuracy of the PeakPicker

methodology.

Previous functional studies carried out with the c.1852_1853AAGC variant have

produced conflicting pieces of evidence regarding its pathogenicity [383], making it difficult to

ascertain if c.1852_1853AAGC is a pathogenic or benign variant. In a previous study using

pulse chase assays and cycloheximide treatments we demonstrated that the

c.1852_1853AAGC variant drastically decreases the stability of the MLH1 protein (Figure

2.6). However, these assays are time consuming and therefore not practical to be applied to the

clinical setting. Furthermore, results from in vitro findings do not always mirror the effect of the

variant in vivo. The results of the PeakPicker analysis demonstrate that this substitution

significantly decreases the expression/stability of the variant allele in the individual‟s

lymphocyte RNA. The IHC findidings indicate that this individual‟s tumour was positive for

MLH1 expression. This may be due to the fact that the variant leads to intermediate levels of

expression, which cannot be accurately determined by a qualitative method such as IHC.

Another possibility is that the mutation that affected the second allele of MLH1 may have

compromised the functionality of the protein without altering its expression (for example by

affecting the ability of MLH1 to bind to PMS2 or to translocate to the nucleus). The differential

effect we see in these two family members could be attributed to unique genetic changes or

environmental exposures that might have influenced the levels of MLH1 transcribed or the

stability of this transcript. The pedigree of this family is depicted in Figure 3.2. However, it is

not possible to rule out if the unbalanced allelic expression profile we observed is the result of a

hitherto undiscovered mutation that lies in regulatory regions or deep within intronic regions that

is in linkage with the variant allele.

Page 148: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

131

The c.2246TC variant was discovered in a proband that developed colorectal cancer at

the age of 24. The pedigree of this proband is depicted in Figure 3.3. This variant lies within the

c-terminal homology domain that is thought to be important in stabilizing interaction between

MLH1 and PMS2[404] and while this variant has been previously reported, no functional studies

have been done to date [411, 412, 435]. Previous studies carried out in our laboratory have

demonstrated that another missense variant affecting the same nucleotide, MLH1 c.2246 TA

did not affect protein expression or stability but showed decreased efficiency of heterodimerizing

with PMS2 [389]. Thus, it is likely that this variant affects heterodimerization of MLH1 rather

than its expression or stability.

Page 149: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

132

Figure 3.2 Pedigree of Case 5A/5B.

The proband in this pedigree was found to carry the unclassified variant K618A. Solid shading

denotes cancer at the time these pedigrees were drawn. The proband is indicated with an arrow.

V+, indicates individuals positive for variant.

Page 150: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

133

Figure 3.3 Pedigree of case 6.

The proband in this pedigree was found to carry the unclassified variant L749P. Solid shading

denotes cancer at the time these pedigrees were drawn. The proband is indicated with an arrow.

Colon dx.24

Liver dx.45

Colon dx.24

Kidney dx.75

Colon dx.24

Page 151: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

134

Truncating mutations are the most common type of mutation identified in both the MLH1 and the

MSH2 genes [436]. Truncating mutations lead to transcripts with premature termination codons

that are generally degraded by nonsense mediated RNA decay, which functions to eliminate

truncated proteins that may have deleterious effects [437]. However, depending on the location

of the termination codon, truncating mutations may or may not lead to decreased mRNA. The

finding that the truncating mutation, c.1528 CT leads to diminished allelic expression/stability,

confirms PeakPicker‟s sensitivity and its versatility in recognizing the effects of diverse types of

genetic alterations.

Unclassified variants in MLH1 have been shown to alter protein expression, stability and

turnover [383, 389]. However, not much attention has been paid to the role of unclassified

variants in leading to allelic imbalances despite its potential to be a noteworthy cause of MMR

deficiency. Allelic imbalances could arise due to genetic alterations that affect the efficiency of

transcription, alter transcript expression or affect mRNA stability and this study provides initial

evidence that a subset of unclassified missense variants in MLH1 can and do affect these

processes. Similarly, a previous study showed that a missense variant in RB1 was able to

significantly impair the allelic balance in a pedigree with retinoblastoma [421]. While the extent

of the effect may depend on the missense alteration in question, it offers another important clue

regarding the pathogenicity of ambiguous variants. Missense variants may alter transcript

expression or stability by leading to incorrect processing or folding of the transcripts.

Another mechanism by which variants may potentially contribute to altered mRNA

expression is through abnormal splicing, and several missense mutations in MLH1 that result in

splicing abnormalities have been shown to be associated with RNA defects [438]. Disruption in

the fidelity of splicing can create unstable mRNAs by activating the NMD system. While none of

Page 152: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

135

the variants used in this study occur in the invariant splice donor and acceptor sites, several other

loosely defined cis-acting sequences have been shown to be important for accurate exon

recognition [429]. Thus, the disruption of these sequences is a likely mechanism by which

unclassified variants may lead to disease susceptibility. An example of this is the c.793C>T

variant, which has been show to lead to exon skipping in several studies However, the splicing

prediction tool, NNSPLICE and the gene prediction program, GeneScan did not predict that

these variants would lead to splicing abnormalities. In addition, previous studies have shown that

pathogenic missense mutations in the MLH1 and MSH2 genes are located in exonic splice

enhancer (ESE) sites significantly more frequently than expected[439]. ESEs are short (6-8 base

pair) degenerate exonic splicing regulatory sequences that serve as the binding sites for

serine/arginine-rich (S/R) proteins. Therefore, alteration of ESEs offers another mechanism by

which these variants may potentially lead to splicing defects. The three in silico approaches we

employed provided dissimilar predictions (Table 3.4). Our analysis indicates that several of the

variants are predicted to lead to the creation, loss and/or the strengthening of putative ESEs.

However, given the redundancy and the complexity of the regulation of splicing, further studies

are required to confirm these predictions as previous studies have indicated that these tools have

limited specificity and sensitivity [429, 430]. Nevertheless, this analysis offers some insight into

the mechanisms that may have led to altered allelic ratios.

A limitation of the PeakPicker strategy is that it is difficult to assay variants that occur

close to exon boundaries or those that occur in relatively small (<80 base pair) exons.

Furthermore, heterozygosity of the variant allele is required to carry out this analysis. However,

these issues can be circumvented if the individual carrying the unclassified variant is

heterozygous for a known, common polymorphism such as c.655A>G. This polymorphism is

suitable for PeakPicker analysis as it has the highest level of heterozygosity among exonic MLH1

Page 153: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

136

polymorphisms [432], and occurs within exon 9 of MLH1 away from intron/exon boundaries. In

this situation, primers flanking the polymorphic region can be used to confirm the effect of the

uncharacterized variant. This is more cost efficient than designing unique primers surrounding

each exon, and can be adopted when a patient population shows a high frequency of a common

SNP. Alternatively in the case of short exonic regions, it may also be possible to use primers

with linker sequences so that useful sequence information is not lost in the process of

sequencing. A short linker sequence was added on to the primers flanking c.350 C>T, which

enabled us to assay this variant, which occurs in a relatively short exon. Importantly, PeakPicker

analysis cannot be used as a stand-alone test, as there may be several variants that do not lead to

allelic imbalances but may possibly affect other functions. Thus, this analysis should be used in

conjunction with a panel of assays that will help determine the putative effect of genetic variants

on expression and/or function. Additionally, this should be combined with other relevant clinical

and family history information indicating tumour MMR deficiency status to obtain a more

accurate understanding of a variant‟s pathogenicity.

Another strength of the PeakPicker analysis is that it will aid in discovering mutations in

individuals that are classified as mutation-negative using conventional diagnostic techniques. A

major problem that arises in the detection of mutations is the possible masking of alterations in

one allele by the normal sequence present on the other allele. Techniques such as conversion

technology and methods based on single nucleotide primer extension are able to analyze the

maternal and paternal alleles separately and have been applied to the detection of MMR

alterations [234, 427, 432]. However, conversion technology is cumbersome and cannot be

readily implemented in the diagnostic setting. Using an allelic expression assay based on single

nucleotide primer extension, Renkonen and colleagues were able to identify the MLH1 R100X

nonsense mutation in a mutation-negative family [427]. PeakPicker analysis, which is very

Page 154: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

137

similar in principal to single nucleotide primer extension assays, offers a very efficient method of

detecting hitherto unidentified genetic changes by measuring imbalances in allelic expression.

Importantly, PeakPicker analysis and the benefits it offers can be extended to the study of other

genes implicated in inherited cancers, as well as other diseases. BRCA1 and BRCA2 are examples

of two such genes that can benefit from this type of analysis as the Breast cancer Information

Core (BIC) database lists about 1500 variants, mostly missense substitutions of unknown clinical

significance [440].

In summary, we demonstrate that the publicly available software tool, PeakPicker was

able to identify MLH1 variants that led to unbalanced expression of mRNA in a time and cost

efficient manner, making it amenable to implementation in the diagnostic setting. Information on

decreased transcript levels, in combination with other assays will help increase the confidence

with which a variant is deemed to be pathogenic, especially if limited clinical data is present.

This technique essentially generates a snapshot of the gene expression profile present in the

individual‟s cells; therefore unlike in vitro assessment strategies this assay takes into

consideration the unique environmental or genetic influences that may be specific to the

individual. The PeakPicker strategy will also open avenues to further analyze mutation-negative

cases. Moreover the strategy described here can also be utilized to help classify genetic

alterations identified in several hereditary diseases, allowing for the improved accuracy and

efficiency of predictive genetic testing programs.

Acknowledgements

We thank the patients and their families for participating in this study. We are grateful to Dr.

Tomi Pastinen (McGill University, Montreal, QC, Canada) and Dr. Bing Ge for providing us

with the PeakPicker software and for helping us set up the program. We also gratefully

Page 155: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

138

acknowledge the assistance provided by the staff of the Molecular Pathology Laboratory, the

Biospecimen Repository and the Familial GI Cancer Registry of the Mt. Sinai Hospital for their

help with this study. In addition, we would like to thank Siwei Chen and the staff of the

Analytical Genetics Technology Centre (University Health Network) for assistance with high

sensitivity sequencing.

Page 156: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

139

CHAPTER 4

Functional Effects of the MLH1-93G>A Polymorphism on MLH1 and

EPM2AIP1 Promoter Activity

Sheron Perera, Miralem Mrkonjic, James B. Rawson and Bharati Bapat.

Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON

Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON

Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON

The work presented in this chapter was primarily contributed by Sheron Perera. MM, a former

Ph.D. student, and JBR, a summer student, assisted in the creation of the promoter constructs.

A modified version of the data presented in this chapter has been submitted for publication.

Page 157: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

140

4 Chapter 4 Functional Effects of the MLH1-93G>A

Polymorphism on MLH1 and EPM2AIP1 Promoter

Activity

4.1 Summary

Defective mismatch repair (MMR) leads to the microsatellite instability (MSI) phenotype of

colorectal cancer (CRC). We have previously shown that the MLH1 c.-93G>A promoter

polymorphism is strongly associated with MSI tumours, suggesting a modifier role for this

polymorphism in CRC. The MLH1 promoter is bi-directional with the EPM2AIP1 gene located

on the antisense strand. To evaluate the functional impact of this polymorphism we transfected a

panel of CRC and endometrial cancer cell lines with luciferase promoter constructs containing

either the -93G or -93A allele. We transfected a core MLH1 promoter construct that spans

nucleotides -301 to +99, and a shorter MLH1 promoter construct that spans nucleotides -113 to

+99. Furthermore, we used a construct in reverse orientation to assess the effect of this

polymorphism on EPM2AIP1. The luciferase activity was compared using a two-sided Student‟s

t-test. Using electrophoretic mobility shift assays (EMSA), we evaluated whether differential

protein binding was responsible for differences in promoter activity. With the core promoter

constructs, we observed higher activity with the -93G allele compared to the -93A allele in

HCT116 colorectal cancer cells. With the shorter promoter constructs, we consistently observed

higher level of activity with the -93G allele in all cell lines examined. This polymorphism also

affected the transcription of the EPM2AIP1 gene. EMSA suggests that this polymorphism alters

the affinity of nuclear factors that bind this region. This indicates that the -93G>A polymorphism

modifies the efficiency of MLH1/EPM2AIP1 transcription.

Page 158: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

141

4.2 Introduction

With over a million cases diagnosed each year, colorectal cancer (CRC) is a significant

cause of morbidity and mortality worldwide. CRCs can be generally subdivided into two

clinically relevant groups depending on their mismatch repair (MMR) status. The DNA

mismatch repair system guards the integrity of the genome and contributes to the overall fidelity

of DNA replication by targeting mispaired bases and insertion-deletion loops that occur through

replication errors, during homologous recombination, and as a result of DNA damage [441].

Defective MMR is also implicated in Lynch syndrome, a cancer predisposition syndrome that

accounts for 2-3% of CRCs. MMR deficiency has also been shown to give rise to sporadic

endometrial cancer with 28% of all endometrial tumours displaying loss of MMR [245].

Moreover, endometrial cancer is also the most common extra-colonic malignancy observed in

Lynch syndrome [442].

Cancers deficient in MMR exhibit genome-wide instability at microsatellite sequences

compared to matched normal tissue DNA. When 30% or more of microsatellite markers tested

show instability, the tumour is defined as high-frequency microsatellite instability or MSI-H.

About 15% of sporadic CRCs and >85% of Lynch syndrome tumours display MSI-H phenotype

[227, 443]. Furthermore, the majority of MSI-H CRCs occur due to the epigenetic silencing of

the MLH1 promoter [444].

Recent evidence has shown the MLH1 promoter to be bi-directional with a second gene,

EPM2AIP1, located in a head-to-head orientation on the opposite strand 321 base pairs away

from the MLH1 transcription start site [342]. Methylation of this promoter region results in

transcriptional silencing of both MLH1 and EPM2AIP1 [342]. Furthermore, MSI-H tumours

show a significant reduction in MLH1/EPM2AIP1 expression compared with normal colonic

Page 159: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

142

mucosa and microsatellite stable (MSS) tumours indicating that EPM2AIP1 expression is turned

off in the majority of sporadic tumours with MLH1 hypermethylation [445]. Although these

findings potentially implicate EPM2AIP1 in CRC, its function is unknown. The MLH1 gene

promoter has been empirically subdivided into four regions based on CpG dinucleotide clusters

and the relationship between their methylation status and MLH1 gene expression. These regions

are termed: A (-711 to -577 base pairs from the MLH1 translation start site), B (-552 to -266), C

(-248 to -178), and D (-109 to +15) [341]. The core promoter region consists of regions C and D,

since methylation of this region is consistently correlated with loss of MLH1 expression in CRC

tumours [339].

A previous genetic epidemiology study carried out in our laboratory aimed at assessing

common single nucleotide polymorphisms identified an association between the MLH1-93G>A

promoter variant and MSI-H CRCs in two separate Canadian populations from the provinces of

Ontario and Newfoundland [446]. Since then these findings have been independently replicated

by other studies as this polymorphism was shown to be associated with increased risk of

hyperplastic polyps and adenomas in smokers [447], as well as MSI-H CRCs [448]. The MLH1-

93G>A variant was also shown to be associated with CRCs that display loss of MLH1 protein

expression [449]. Furthermore, the MLH1-93G>A variant was shown to increase the risk of

developing endometrial cancer [450]. The correlation between the -93G>A SNP and cancer has

not been limited to CRC or endometrial cancer, with previous studies reporting an association

with increased risk of ovarian [451] and squamous cell lung cancer [452]. Taken together, these

studies provide evidence that the -93G>A variant is likely to mediate functional effects

promoting carcinogenesis. However, there has been no systematic investigation of the functional

contribution of this variant to date, and the limited data that exists are conflicting [453, 454].

Furthermore, no study has evaluated cell-type specific influences of this variant, which are

Page 160: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

143

important given differences in transcription factor expression and regulation in diverse host cell

types [455].

We carried out a comprehensive investigation of the role of the -93G>A variant in

altering MLH1 transcription in a subset of cell lines representative of colorectal and endometrial

cancers. In addition, we extended our analysis to determine if this variant affects transcription of

the EPM2AIP1 gene, in order to elucidate a role in gene in carcinogenesis.

4.3 Materials and Methods

4.3.1 Generation of Promoter Constructs

pGL3-Basic plasmids containing the C+D region (-301 to +99) of the MLH1 promoter

(ENST00000231790) were gifts from Dr. Hiromichi Hemmi (Toho University School of

Medicine, Japan). These plasmids, which contain either the G or A nucleotide at the -93 location,

have been described previously [454].

To generate plasmids of the MLH1 promoter D-region with the G or A allele, DNA

fragments corresponding to the MLH1 promoter D region (-113 to +99) were PCR amplified

from the appropriate C+D promoter constructs. The PCR reaction contained 10X Taq Buffer

HiFi, 2 mM MgCl2, 0.2 mM dNTPs, 1 mM primers, 14 ng template DNA, and 1 unit high-

fidelity Taq DNA polymerase (Fermentas Life Sciences, Burlington, ON, Canada). The PCR

conditions were as follows: initial denaturation (95 C for 10 minutes), denaturation( 95 C for 30

seconds), annealing (57 C for 45 sec), extension (72 C for 1 min), and final extension (72 C for

8 min). The denaturation-annealing-extension steps were repeated 35 times. The 5‟ to 3‟

sequences of the forward and reverse primers, respectively, are:

Page 161: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

144

AAAAAAAACTCGAGGGATGGCGTAAGCTA (XhoI site underlined),

AAAAAAAAAAGCTTCTTTGATAGCATTAGCTGGCCG (HindIII site underlined).

To generate plasmids with the EPM2AIP1 promoter G or A allele, DNA fragments

corresponding to the -513 to +122 region of the MLH1/EPM2AIP1 promoter were amplified

from lymphocyte DNA of CRC patients homozygous (for G or A) at the -93 locus. The PCR

conditions were identical to that described for the MLH1 promoter D-region except that 40 ng of

template DNA and 2.5 mM MgCl2 were used. The 5‟ to 3‟ sequences of the forward and reverse

primers, respectively, are: CCTCGTCGACTTCCATCTTGCTTCTTTT and CCGTACCAGT

TCTCAATCATCTCTTTGAT. A second, nested PCR was performed using the PCR fragment

generated above as templates. The PCR conditions were identical to the above reaction except

that 20 ng of template DNA was used. The 5‟ to 3‟ sequences of the forward and reverse primers,

respectively, are: AAAAAAAAAAGCTTCACAAGCCCGGTTCC (HindIII site underlined) and

AAAAAAAACTCGAGAAACGTCTAGATGCTCAACGG (XhoI site underlined). Following

all PCR reactions, DNA was isolated by gel electrophoresis and purified by QIAquick Gel

Extraction Kit (Qiagen, Mississauga, ON, Canada). MLH1 D-region promoter constructs,

EPM2AIP1 promoter constructs, and pGL3-Basic vector were digested with XhoI and HindIII

restriction endonucleases (Fermentas). The restriction digestion reaction used 10X Buffer R with

BSA, 12.5 units of each restriction enzyme, and a 3:1 insert to vector ratio. The reaction mix was

incubated at 37 C for 18 hrs. A second round of restriction enzymes was added at 8 hrs to ensure

maximum digestion. Following the digestion, 5 l of shrimp alkaline phosphatase (SAP) was

added to prevent religation of the plasmid and the mixes were incubated at 37 C for 1 hr.

Following enzyme inactivation at 80 C for 20 min, DNA was purified using QIAQuick PCR

Purification Kit (Qiagen). The integrity of all constructs was verified by DNA sequencing.

Page 162: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

145

Figure 4.1 The schematic of the MLH1/EPM2AIP1 bi-directional promoter.

The MLH1 promoter is empirically divided into four regions A-D as shown [341]. The

nucleotides demarcating the start and end of each region are indicated (numbering is with

reference to the MLH1 translational start site). The core promoter region, which comprises the C

and D regions, is shared between the MLH1 and EPM2AIP1 genes. The translational start sites of

MLH1 and EPM2AIP1 are shown with arrows. The promoter constructs used in luciferase

reporter assays are indicated below. The C+D construct extends from -301 to +99, the D

construct from -113 to +99 and the EPM2AIP1 construct from -513 to+122. The asterisk

represents the -93G>A polymorphism located in D-region. The figure is not drawn to scale.

Page 163: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

146

4.3.2 Cell Culture

Tissue culture reagents and fetal bovine serum (FBS) were purchased from Invitrogen

Life Technologies (Burlington, ON, Canada). HCT116 and HEC-1-A cells were cultured in

McCoy‟s 5A media. SK-UT-1B was maintained in Eagle‟s minimum essential medium (ATCC,

Manassas, VA). All cell culture media was supplemented with 10% FBS. All cell lines were

incubated at 37 C in a humidified atmosphere of 5% CO2.

4.3.3 Luciferase Reporter Gene Assays

All transfection experiments were carried out using Lipofectamine 2000 reagent

(Invitrogen Life Technologies) according to manufacturer‟s instructions. Cells were plated in 24-

well plates at a density of 100,000 cells per well 24 hours prior to transfection. The promoter-less

pGL3-Basic (empty vector) was used as a negative control. Luciferase constructs (500 ng)

containing either pGL3-Basic, MLH1 C+D promoter constructs (-93G or A), MLH1 D promoter

constructs (-93G or A), or EPM2AIP1 promoter constructs (-93G or A), were transfected into

cells to determine luciferase reporter activity [454, 456]. Cells were additionally co-transfected

with an internal control (5 ng of pRL-SV40, Promega, Madison, WI) for monitoring transfection

efficiency and normalization. Luciferase and Renilla activities were measured 24 hr after

transfection using the Dual Luciferase Reporter Assay System (Promega), which allows the

firefly and Renilla luciferase activities to be quantified in the same sample. Luminescence was

quantified using a Berthold 96-well microplate luminometer (Berthold, Wildbad, Germany). All

reporter assays were done at least in triplicate, with at least three independent experiments

performed. Statistically significant differences in promoter activity were assessed using a two

sided Student‟s t-test. Error bars represent standard deviation of the mean (SD).

Page 164: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

147

4.3.4 Western blotting

Cold radioimmunoprecipitation assay (RIPA) lysis buffer (0.5 % Nonidet P-40, 0.5% sodium

deoxycholate, 0.1% sodium dodecyl sulfate, 1x PBS) buffer containing Complete protease

inhibitor tablets (Roche, Mannheim, Germany) was used to lyse the cells on ice for

approximately 20 minutes. The lysates were then collected by centrifugation at 14,000 rpm for

approximately 15 minutes. The expression levels for the proteins were examined by sodium

dodecyl sulphate/ polyacrylamide gel electrophoresis, followed by western blot analysis with

anti-MLH1 (clone 168-728; BD Biosciences) and anti-EPM2AIP1 (developed by Julie Turnbull

and Dr. Berge Minassian, University of Toronto)

4.3.5 Electrophoretic Mobility Shift Assays (EMSA)

The following double stranded probes (variant shown in bold within brackets) that were

labeled with biotin on the 5‟ end were purchased from Integrated DNA Technologies (Coralville,

IA) : -93G sense, 5‟-TAAGCTACAGCT[G]AAGGAAGAACGTG-3‟; -93G antisense,

5‟CACGTTCTTCCTT[C]AGCTGTAGCTTA-3‟; -93A sense,

TAAGCTACAGCT[A]AAGGAAGAACGTG-3‟; -93A antisense, 5‟-

CACGTTCTTCCTT[T]AGCTGTAGCTTA-3‟. The probes generated for the competition

reaction were identical, except they did not contain a 5‟ biotin group.

Nuclear extracts of the cell lines were prepared using the NE-PER kit (Pierce, Rockford,

IL) according to the manufacturer‟s instructions. EMSA was carried out using the LightShift

Chemiluminescent EMSA Kit (Thermo-Fisher, Rockford, IL) according to the instructions of the

manufacturer. We incubated 15 µg of nuclear proteins from the HCT116 cell line and 8 µg of

nuclear protein from the HEC-1-A cell line with 60 fmol biotin-labeled oligonucleotide for 25

min at room temperature in binding buffer (10 mM Tris, 50 Mm KCl, 1 mM dithiothreitol, pH

Page 165: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

148

7.5), 50 ng/µl poly (dI-dC), 0.05% NP40, and 3.125% glycerol. Binding was competed by 200,

400, 800 and 2000-fold excess unlabeled oligonucleotides. Binding complexes were resolved by

electrophoresis using 5% TBE Criterion gels (Bio-Rad, Hercules, CA), transferred to Biodyne B

pre-cut modified nylon membranes (Pierce), UV cross-linked, and visualized

using the

Chemiluminescent Nucleic Acid Detection system (Pierce). The EMSAs were performed at least

at least three times independently.

Supershift experiments were conducted using specific antibodies (rabbit anti-DEC2 and anti-

Max Santa Cruz Biotechnology, Santa Cruz, CA). Reactions were identical to gel shift reaction

conditions except 2μg or 4μg of antibody along with the binding reaction mixtures was incubated

for 60 minutes on ice before the addition of labeled probe. Once the probe was added the

reaction mixtures were incubated for 25min at room temperature prior to electrophoresis as

described above.

4.4 Results

4.4.1 Effects of the MLH1 -93G>A variant on promoter activity in colorectal

cancer cell lines

We examined the effects of the MLH1-93G>A promoter polymorphism on MLH1

promoter activity using two different constructs, one that spans a core promoter C+D region (-

301 to +99) and another that spans only the shorter D region of the promoter (-113 to +99;

Figure 4.1). In the HCT116 cell line, the -93G allele shows ~1.28 fold higher transcriptional

activity of MLH1 compared to the -93A allele when transfected with the core promoter construct

(P = 0.0009; Figure 4.2). When the D construct was transfected, the activity of the -93G allele

was ~5 fold higher than the -93A allele (P = 0.0025). Promoter activities of all constructs are

summarized in Table 4.1.

Page 166: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

149

4.4.2 Effects of the MLH1 -93G>A variant on promoter activity in endometrial

cancer cell lines

We assessed the effect of this polymorphism on the transcriptional activity of MLH1 in

the human endometrial cancer cell lines, HEC-1-A and SK-UT-1B (Figure 4.3). For both cell

lines, there was no significant difference in luciferase reporter activity between the two alleles

when the core promoter construct containing the C+D region was used (Figure 4.3). However,

with the D region promoter construct, the -93G allele showed a 4.3 fold (P = 0.0005) and a 9.5

fold (P = 0.001) higher activity compared to the -93A allele in HEC-1A and SK-UT-1B

respectively (Table 4.1).

4.4.3 Characterization of MLH1 and EPM2AIP1 in a panel of human cell lines

EPM2AIP1 shares the same promoter with MLH1. However, neither the function of this gene nor

its expression status in colon and endometrial cancer is known. As a preliminary analysis, we

determined the expression of EPM2AIP1 in a panel of human cell lines derived from cancers of

different sites, as well a non-tumourigenic cell line. Our results suggest that EPM2AIP1 protein

is expressed in a range of cell lines as a doublet (Figure 4.4). Furthermore, this expression

correlates with the mutation/methylation status of MLH1 in these cell lines. HCT116 cells have a

hemizygous nonsense mutation [414], thus its deficient for endogenous MLH1, but expression of

EPM2AIP1 is not affected. The SW620 cell line is neither methylated nor mutated (Unpublished

observations, Dr. Richard Hamelin, INSERM, France); therefore both MLH1 and EPM2AIP1

are expressed. The shared MLH1/EPM2AIP1 promoter is hypermethylated in the embryonic

kidney cell line, HEK293T [385] making it deficient for both of the proteins. The prostate,

endometrial and breast cancer cell lines (PC3, SK-UT-1B and MCF-7 respectively) appear to

show expression of both MLH1 and EPM2AIP1.

Page 167: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

150

4.4.4 Effects of the MLH1 -93G>A variant on EPM2AIP1 promoter activity

Because of the bi-directional nature of the MLH1/EPM2AIP1 promoter [342], we

evaluated the effects of the -93G>A polymorphism using the reverse promoter constructs (Figure

4.1). In the HCT116 (P = 0.007) colorectal cancer cell line the -93G allele showed a statistically

significant decrease in promoter activity compared to the -93A allele (Table 4.1, Figure 4.5).

In the endometrial cancer cell line HEC-1-A the -93G allele displayed lower activity than

the -93A allele (P = 0.004), similar to the colorectal cancer cell lines. However, in the SK-UT-

1B cell line this trend was reversed, with the -93G allele showing statistically significantly

higher activity(P = 0.024, Figure 4.5).

Page 168: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

151

C D

Page 169: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

152

Figure 4.2 Effect of the MLH1-93G>A polymorphism on MLH1 transcriptional activity in

the HCT116 cell line.

Effects of the -93G>A polymorphism were evaluated using luciferase constructs encompassing

the C+D region (A, C) or the D region (B, D) containing either the G or A allele at the -93

location. Results shown for the -93 G or -93A allele were derived from firefly luciferase activity

normalized to Renilla, and expressed relative to the EV plasmid. Each sample was assayed at

least in triplicate, with three independent experiments performed. Statistically significant

differences in promoter activity were assessed using the Student‟s t-test and are indicated by an

asterisk. In bar graphs error bars represent standard deviation from the mean, and in dot plots

error bars represent 95% confidence interval. EV: empty vector (pGL3-Basic) construct, C+D:

construct containing the C and D region of MLH1 promoter, D: construct containing the D region

of MLH1 promoter

Page 170: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

153

Page 171: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

154

Figure 4.3 Effect of the MLH1-93G>A polymorphism on MLH1 transcriptional activity in

the endometrial cancer cell lines.

Luciferase constructs containing either the G or A allele at the -93 encompassing the C+D region

or the D region were transfected into HEC-1-A (A, B, E, F) and SK-UT-1B (C, D, G, H) cell

lines. Results shown for the -93G or -93A allele were derived from firefly luciferase activity

normalized to Renilla, and expressed relative to the EV plasmid. Each sample was assayed at

least in triplicate, with three independent experiments performed. Statistically significant

differences in promoter activity are indicated by an asterisk. In bar graphs error bars represent

standard deviation from the mean and in the dot plots error bars represent the 95% confidence

interval.

Page 172: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

155

Figure 4.4 Expression profile of endogenous MLH1 and EPM2AIP1 in a panel of human

cell lines.

Total extracts were immunoblotted with either anti-MLH1 (upper panel) or anti-EPM2AIP1

(lower panel) (Note that the blot was cut in half and then immunoblotted for the two proteins).

HCT116 cells have a hemizygous nonsense mutation[414], thus its deficient for endogenous

MLH1, but expression of EPM2AIP1 is not affected. The SW620 cell line is neither methylated

nor mutated (Unpublished observations Dr. Richard Hamelin); therefore both MLH1 and

EPM2AIP1 are expressed. The shared MLH1/EPM2AIP1 promoter is hypermethylated in the

HEK293T cell line [385], making it deficient for both of the proteins. The prostate, endometrial

and breast cancer cell lines (PC3, SK-UT-1B and MCF-7 respectively) appear to show

expression of both MLH1 and EPM2AIP1.

Page 173: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

156

Page 174: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

157

Figure 4.5 Effect of the MLH1-93G>A polymorphism on EPM2AIP1 transcriptional

activity.

Constructs that span the shared bidirectional promoter, which contain either the A or G

nucleotide at the -93 position (relative to MLH1) were transfected into HCT116 (A, D), HEC-1-

A (B, E), and SK-UT-1B (C, F). Results shown for the -93 G or A allele were derived from

firefly luciferase activity normalized to Renilla, and expressed relative to the EV plasmid. Each

sample was assayed at least in triplicate, with three independent experiments performed.

Statistically significant differences in promoter activity are indicated by an asterisk. In bar graphs

error bars represent standard deviation from the mean and in dot plots error bars represent the

95% confidence interval.

Page 175: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

158

Table 4.1 Summary of promoter activities

MLH1 promoter activities

Cell Line Construct Relative Luciferase Activity

P-

value

-93G ± SD 95% CI -93A ± SD 95% CI

HCT116 C+D 90.38± 2.99 87-93.76 70.37±2.15 67.94-72.80 0.001

D 69.38±4.32 64.49-74.27 13.81±0.59 13.14-14.48 0.002

HEC-1-A C+D 138.08±10.11 126.64-149.52 144.59±18.37 123.80-165.38 0.470

D 15.85±0.34 15.47-16.23 3.69±0.16 3.51-3.87 0.001

SK-UT-

1B

C+D 100.45±11.21 87.76-113.14 89.15±7.12 81.09-97.21 0.335

D 83.52±2.45 80.75-86.29 8.8±1.56 7.03-10.57 0.001

EPM2AIP1 promoter activities

Cell Line Relative Luciferase Activity P-

value

-93G ± SD 95% CI -93A ± SD 95% CI

HCT116 139.73±7.39 131.37-148.09 222.73±7.05 214.75-230.71 0.007

HEC-1-A 26.94±1.14 25.65-28.23 34.77±1.38 33.21-36.33 0.004

SK-UT-1B 255.15±11.83 241.76-268.54 161.52±37.19 119.44-203.60 0.024

n=3 independent experiments with each samples assayed at least in triplicate. P –values are

calculated using a two-sided Student‟s t-test. SD: standard deviation, CI: confidence interval.

Page 176: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

159

4.4.5 Effect of the MLH1 -93G>A promoter variant on transcription factor

binding

We carried out EMSA to assess whether the differences observed in the luciferase

reporter activity between the -93G and the -93A allele were due to differential binding of nuclear

factors. These experiments revealed that multiple factors bound the probe. We used increasing

amounts of unlabelled probe as a competitor to verify the specificity of binding interactions. In

reactions carried out with the nuclear extract of the HCT116 cell line, multiple factors bound to

the labeled probe (Figure 4.6). In general, the specificity appeared to be very high with 800-

2000x unlabelled probe required to compete out the binding reaction. In addition, these factors

appear to have different affinities for the binding sequence and consequently seem to exhibit

competitive binding. Addition of the unlabelled competitor probe diminished certain interactions

and strengthened others. When the binding patterns with the -93G probe were compared to that

of the -93A probe, we observed that one of the factors (arrowhead, Figure 4.6) had higher

affinity for the -93G probe. Furthermore, the highest concentration of competitor was unable to

compete out binding with this factor, while this was not so with the -93A allele.

To investigate the effect of this polymorphism on a different cell type, EMSA was also

performed with nuclear extract from the HEC-1-A cell line (Figure 4.7). Here too, we observed

the binding of multiple factors to the probe, as well as competition between the different factors

that bound the region. One nuclear factor (arrowhead, Figure 4.7) appeared to have higher

affinity for the -93A allele, as it remained bound even in the presence of 2000x unlabelled probe,

while with the -93G probe 800x unlabelled probe was sufficient to compete out this reaction. We

also observed that the -93A unlabelled probe was unable to compete out the reaction with the -

93G labeled probe (and vice versa), which provided further evidence for the specificity of these

interactions.

Page 177: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

160

Figure 4.6 Impact of the MLH1 -93 >A polymorphism on nuclear factor binding in

HCT116.

At least 3 independent EMSAs were carried out to determine the effects on binding and a

representative experiment is depicted. Unlabelled competitor was added in decreasing

concentrations (2000x, 800x and 400x respectively) to verify the specificity and the affinity of

the interactions. (+) reactions carried out in the presence of nuclear lysate; (-) reactions carried

out without nuclear extract or without competitor. The arrowhead highlights factor(s) that appear

to show differential binding between the -93A and -93G alleles.

Page 178: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

161

Figure 4.7 Impact of the MLH1 -93 >A polymorphism on nuclear factor binding in HEC-1-

A.

At least 3 independent EMSAs were carried out to determine the effects on binding and a

representative experiment in depicted. Unlabelled competitor was added in decreasing

concentrations (2000x, 800x and 400x respectively) to verify the specificity and the affinity of

the interactions. (+) reactions carried out in the presence of nuclear lysate; (-) reactions carried

out without nuclear extract or without competitor. The arrowhead highlights factor(s) that show

differential binding between the -93A and -93G alleles.

Page 179: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

162

Page 180: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

163

Figure 8 Figure 4.8 Determination of putative factors that bind the promoter region

surrounding -93G>A.

Max and DEC2 antibodies were added to the binding region to determine if these transcription

factors bound the region surrounding the variant in nuclear lysates from the HEC-1-A cell line

(A, B) and the HCT116 cell line (C). Unlabelled competitor was added to verify the specificity

and the affinity of the interactions. (+) reactions carried out in the presence of nuclear

lysate/unlabelled competitor; (-) reactions carried out without nuclear extract or without

competitor. (1): 2µg of Max antibody, (2): 4µg of Max antibody, (3): 2µg of DEC2 antibody, (4):

4µg of DEC2 antibody.

Page 181: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

164

Max is a member of the basic helix-loop-helix leucine zipper (bHLHZ) family of transcription

factors and forms homodimers and heterodimer with other family members. DEC2 is a basic-

helix-loop-helix (bHLH) transcription factor and both proteins have been implicated in the

regulation of MLH1. In an attempt to shed light on the identity of the factors that bind this region

we carried out supershift assays with 2µg or 4µg of anti-DEC2 or anti-Max antibody in HEC-1-

A and HCT116 nuclear lysate (Figure 4.8). In these assays we did not observe a supershift or an

interference of the specific bands indicating that these factors are not involved in binding this

region in these cell types.

4.5 Discussion

In this study, using constructs of two different lengths spanning the MLH1/EPM2AIP1

promoter region, we were able to show that the -93 G>A promoter polymorphism affects the

transcription of MLH1 in cell lines derived from cancers of the colorectum and endometrium.

Similar effects were also seen in cell lines derived from non-tumourigenic tissues as well

(unpublished data included in manuscript that is currently under review, experiments performed

by Miralem Mrkonjic, a former PhD student in our laboratory). In the CRC cell line HCT116; we

saw that the -93G allele was significantly more active than the -93A allele when the core

promoter (C+D) construct was examined. We consistently observed increased promoter activity

for the -93G allele with the D constructs in the three cell lines examined. Similar results were

observed in several other CRC cell lines as well (unpublished data, Miralem Mrkonjic). Thus,

the effect of the MLH1 -93G>A polymorphism on promoter activity was much more pronounced

in the shorter promoter constructs. This was likely due to its abbreviated length, as the longer

Page 182: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

165

core promoter construct likely harbors multiple transcription factor recognition sites that might

eclipse the subtle effect of the -93G>A polymorphism. As expected, the overall activities of the

D-region promoter constructs were considerably lower compared to the core promoter

constructs. This is likely due to differential expression and regulation of transcription factors. A

universal dramatic effect on the transcription of MLH1 at the level of the core promoter would

likely have a larger impact on the associated cancer risk. If the -93G>A were to have such a

detrimental effect, then it would likely be categorized as a disease causing mutation rather than a

polymorphism. This is supported by the fact that about 23% of the general, unaffected,

population are carriers of this variant. Therefore, our finding that this polymorphism has a subtle

but significant and consistent effect at the level of the minimum D promoter region is more in

line with the epidemiological data.

In addition to its role in MLH1 transcription, analysis of the reverse promoter constructs

indicate that the -93G>A promoter polymorphism also plays a role in transcription of the

EPM2AIP1 gene. Our colorectal and endometrial cancer cell lines demonstrated differences in

the level of EPM2AIP1 transcribed by the -93G and -93A alleles. However, in two of the cell

lines, HCT116 and HEC-1-A the -93A allele demonstrated higher activity. However, in SK-UT-

1B, there was a significant decrease in the activity of the -93A allele. This discrepancy could

have arisen due to the cell specific expression of factors involved in transcription. These results

indicate that while the overall effect of this variant on the transcription of EPM2AIP1 may differ

in individuals and/or cell types, the presence of this polymorphism alters the level of EPM2AIP1

transcribed. Interestingly, the -93G>A polymorphism is located in a region that was previously

shown to be crucial for EPM2AIP1 transcription and promoter methylation that silences the

MLH1 gene also results in loss of EPM2AIP1 gene expression [342]. Nonetheless, the

function(s) of the EPM2AIP1 is currently unknown [343]. A genome-wide breast cancer

Page 183: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

166

association study identified EPM2AIP1 gene as a high-priority candidate that modifies breast

cancer risk in BRCA2 mutation carriers [457]. EPM2AIP1 was found to interact with Laforin

(EPM2A), which was implicated in the Wnt signaling pathway as well as rapid onset

tumourigenesis in mice [458]. Nevertheless, it is unclear whether EPM2AIP1 plays a role in

Laforin-mediated tumourigenesis. Our results offer evidence that genetic alterations in the

promoter region of MLH1, hitherto considered only for their potential effect on MLH1

transcription can potentially regulate the transcription of EPM2AIP1 as well.

Given the observed effect of the -93G>A variant on both the MLH1 and EPM2AIP1

genes in CRC and endometrial cancer cell lines, it is important to assess how this variant might

influence transcription. One possible mechanism is by altering transcription factor binding sites.

Our EMSA results indicate that the region surrounding the -93G>A variant was bound by

multiple nuclear factors and that the -93G>A variant altered the affinity and binding pattern of

these factors in both CRC, endometrial and non-tumourigenic cell lines. EMSA allows for the

assessment of the abundance, relative affinity and binding specificity of DNA-binding

proteins[459]. These results corroborate the findings of our promoter activity assays. However,

our experimental design cannot determine the identity of the nuclear factors. Several studies have

analyzed the promoter region of MLH1 for potential transcription factor binding sites. One study

identified a protein-binding site at the -93G>A location, which suggests that the region

surrounding the variant is important for protein binding. However, no known proteins were

predicted to bind this region according to the TFSEARCH and DNASIS software [456]. While

knowing the identity of the factors will help shed further light on the mechanism by which this

promoter polymorphism mediates its effects on gene transcription, identifying these factor(s) will

not necessarily help interpret the risk associated with this variant. Given the different binding

patterns observed between the colorectal, endometrial and normal colonic cell lines, it is possible

Page 184: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

167

that considerable variation exists in the nuclear factors that bind this region between different

host cell lines. The results from our EMSA provide further evidence that the -93G>A variant

alters transcription of the two genes, MLH1 and EPM2AIP1, that are regulated by a common

promoter.

Two proteins implicated in binding the MLH1 promoter are DEC2 and Max. The

transcription factor Max has been shown to interact with MSH2 both in vitro and in vivo [460].

Recent studies implicated Max in repressing MLH1 and MSH2 under hypoxic conditions. This

repression was shown to occur via a dynamic shift in occupancy from activating c-Myc/Max to

repressive Mad1/Max and Mnt/Max complexes at the proximal promoters of both the MLH1 and

MSH2 genes [461]. These factors recognize E-Box motifs in the DNA. The hypoxia-inducible

transcription repressor, DEC2 has been shown to regulate the transcription of MLH1 by binding

to E-box-like motif(s) in the MLH1 promoter region [462]. While little is known about the

microenvironmental factors that may lead to genetic instability in the tumour microenvironment,

emerging evidence suggests that hypoxia plays a key role in this process. It was shown that

hypoxia downregulates the expression of the MLH1 gene at the transcriptional level [463]. An E-

Box motif (AACGTG), is located 8 nucleotides downstream from the -93G>A polymorphism.

However, results from our supershift assays did not provide evidence of DEC2 or Max as

possible factors that bound the region surround the -93 variant of the MLH1 promoter. However,

it is possible that these interactions occur only under hypoxic conditions or that these interactions

are specific to certain cell types. Our analysis does not exclude the possibility of these

interactions as it could occur outside of the 25 base pair region spanning our probe.

The main limitation of our study is our inability to conclude whether the proteins that

bind the region surrounding the -93G>A variant in the EMSA experiments are activators or

Page 185: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

168

repressors. Neither are we able to specifically delineate if these factors regulate MLH1,

EPM2AIP1, or both genes. In addition, given our observation that multiple factors bind the

labeled probe, it is unclear whether a subset of the observed proteins represents multi-protein

complexes of coactivators or corepressors, or distinct transcription factors.

A previous study found the -93G>A variant to be associated with MLH1 methylation in

endometrial and colorectal tumours [464, 465], which raises the possibility that this

polymorphism affects binding of methylation machinery and therefore results in gene silencing.

Thus, in addition to modifying the binding of transcription factors, this polymorphism appears to

be able to alter promoter methylation in a context dependent manner. Furthermore, it is likely

that these effects are not necessarily independent of each other and that they act in conjunction in

certain instances. For example, differential methylation of the MLH1 promoter has been

observed in CRC [339, 466]; however, differences in methylation between the C and D regions

have not been studied [340]. Hypothetically, if a large part of the MLH1 promoter C region is

methylated, then the role of the D region in mediating transcription might be enhanced, leading

to a greater effect of the -93G>A variant. Biologically, this effect could occur in individuals

whose MLH1 promoter is partially methylated leaving the C-region inactivated, while the

promoter D-region remains unaffected.

This study has several strengths. This is the first study to examine the effects of the

MLH1-93G>A promoter polymorphism on promoter activity in MLH1 promoter constructs of

two different lengths. In addition, we demonstrate the functional effects of this polymorphism in

a panel of cell lines representative of colorectal and endometrial cancer. This is also the first

study to evaluate the effects of this variant on transcription of EPM2AIP1, demonstrating that a

genetic alteration simultaneously affects both the regulation of MLH1 and its antisense gene. We

Page 186: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

169

demonstrate that MLH1 -93G>A is a functional polymorphism in which the A allele results in

decreased MLH1 transcription. This builds on our previous finding that the -93A allele is

associated MSI-H CRCs, and in combination with previously published genetic epidemiology

studies, supports its role in modifying the risk of cancer development. Further functional

characterization of risk alleles identified in population-based studies will enhance our

understanding of cancer initiation and progression and may possibly serve as potential tools for

screening and prognosis.

Acknowledgements

We would like to thank Julie Turnbull and Dr. Berge Minassian (Hospital for Sick Children,

Toronto) for sharing with us the anti-EPM2AIP1 antibody that they generated.u

Page 187: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

170

CHAPTER 5

5 Discussion and Future directions

Germline mutations in the MMR genes lead to Lynch syndrome and genetic testing is offered to

identify individuals at risk of developing this disease. About a third of all genetic alterations

detected in the mismatch repair gene MLH1 are missense variants, the majority of which are

variants of unclassified clinical significance. In this thesis, our overall aim was to investigate the

mechanism(s) by which a panel of missense variants localized to distinct domains impacted the

regulation, expression and function(s) of MLH1.

5.1 Stability and dimerization of MLH1

The work presented in this thesis demonstrates for the first time that two missense substitutions,

p.R265C and p.K618A alter the stability of the MLH1 protein. Although altered stability has

been proposed as mechanism by which unclassified variants affect MLH1 expression, this had

not been definitively demonstrated. However, the pathway responsible for the degradation of

MLH1 is currently unknown and as such it is difficult to explain the mechanism responsible for

this observed decrease in stability. Based on our findings, we speculate that missense

substitutions may affect the tertiary structure of the protein, which may lead to defective folding

and the consequent degradation of MLH1. It can be postulated that irrespective of the

functionality of the protein, decreased stability likely leads to a situation where lower levels of

protein are present in the cell. Although the levels of protein essential for normal cellular

function and the threshold below which cancer susceptibility occurs has not been established; it

is likely that decreased expression compromises to some degree function(s) carried out by these

Page 188: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

171

protein complexes. It is also possible that varying threshold levels of MLH1 exist for the

different functions of the MMR system, even in the same cells or tissues. This was underscored

in a previous study that indicated that less MLH1 expression is required for the post-replicative

error repair activity of MLH1 compared to the DNA damage response function[311]. This

illustrates how these variants may potentially impact one function, while leaving another intact.

If this situation holds true in Lynch syndrome patients, it will explain how genetic variants not

associated with the MSI-H phenotype in tumours (a surrogate marker of defective post-

replicative error repair) could potentially lead to disease susceptibility.

Our findings also offer some insight into the stability of wild-type MLH1. However, the

mechanism responsible for MLH1 is degradation and turnover is currently unknown. Given that

the sustained expression of MLH1 is required to stabilize the expression of the other MutL

homologs [467], further studies regarding the mechanism responsible for MLH1 degradation is

warranted. These studies will likely shed light on the mechanisms by which MLH1 is regulated,

a topic on which currently very little is known.

Another unclassified variant, p.L749Q impacted the ability of MLH1 to heterodimerize with

PMS2. All MMR functions described to date require that the MutL homologs be localized to the

nucleus of the cell. Furthermore, it has been shown that the dimerization of MLH1 and PMS2 is

necessary for the nuclear localization of MutLα [336]. Additionally, a recent study found that

the nuclear localization of MLH3 is dependent on MLH1, and is competitive with PMS2 [468,

469]. This raises the possibility that missense variants in MLH1 that alter its ability to bind to

specific MutL homologs, alter the relative abundance of MutL complexes in the cells and in

doing so alter the functional capabilities of this system.

Page 189: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

172

5.2 The role of MLH1 in DNA damage signaling

Using a stable inducible cell line model, we were not able to ascertain if MLH1 variants alter

MLH1‟s ability to signal DNA damage. However, in spite of these technical setbacks this

remains an important question to be addressed. Numerous studies have linked MLH1 and the

MMR proteins as important players in signaling DNA damage caused by a range of agents.

However, the precise mechanism by which this occurs remains to be resolved. Gross genetic and

or biochemical approaches such as knocking down the entire MLH1 protein might interfere with

more than function of the MMR system, leading to potentially confounding findings. The ability

of missense variants in MSH2 to uncouple the functions of the MMR system [381], underscores

the utility of missense alterations in evaluating the diverse functions of the MMR proteins. The

lack of functionality associated with MLH1 overexpression in our cell line system demonstrates

that increased expression, like decreased expression can potentially be harmful to the cell. Given

our experiences, in future studies it will be prudent to generate cell lines carrying bacterial

artificial chromosomes (BAC) of wild-type and variant MLH1. BACs are useful as it maintains

not only the coding sequence but also the regulatory sequences upstream of it, including various

promoter sequences that may potentially govern a gene's expression level. Given that the level of

MLH1 appears to be maintained at a delicate balance, a system of this nature will minimize the

potentially negative effects of artificially altered expression. If this approach identifies variants

that are associated with cell cycle arrest or apoptosis, then microarray analysis can be performed

on these cell lines to determine the panel of genes that are up- or down-regulated in these cells.

The benefit of this experimental system would be the ability to compare two, otherwise identical

cell lines that express a protein that differ by only one amino acid, which should at least

theoretically yield a smaller subset of candidate genes than comparing two independent cell

Page 190: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

173

lines. It is expected that this will help identify novel regulators of the MMR mediated DNA

damage signaling pathway. Moreover, if this approach shows interesting results, then mouse

models of these variants can be generated and their phenotypes‟ can be compared to Mlh1-/-

and

Mlh1+/-

mice. The studies that demonstrated the uncoupling of the DNA damage signaling

function from that of the post-replicative error repair function was carried out in this manner

using mice that carry the G674A missense mutation [381]. In addition, ES cells from these mice

can be used to both investigate the presence of genetic instability, as well as perform experiments

evaluating the functional consequences of these variants under controlled but physiologically

relevant conditions.

Moreover, the majority of the efforts to date that aim to characterize unclassified variants in

MLH1 have focused on the post-replicative error repair function. While this is undoubtedly a

principle function of the MMR proteins, in the face of emerging evidence, it is unlikely to be the

only mechanism by which variants mediate their pathogenicity. However, at present there are no

widely-applicable and clinically relevant ways of assessing the impact on these functions. A

better understanding of the role of MLH1 and the MMR system in responding to DNA damage

can be utilized to develop relevant functional assays. A recent study described a highthroughput

system for generating BAC transgenes and stably transfecting them into cell lines [470]. If such a

method can be adopted, then this will allow for multiple variants to be screened simultaneously.

Furthermore, a better understanding of the role of MLH1 in signaling DNA damage will help

develop anti-cancer treatments that exploit the MMR expression status of a tumour.

Page 191: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

174

5.4 Allelic imbalances in MLH1 and its implications for predictive genetic

testing programs

The work presented in this thesis also demonstrates that a subset of missense variants in MLH1

lead to allelic imbalances. Allelic imbalances could arise due to genetic alterations that affect the

efficiency of transcription, alter transcript expression and/or affect mRNA stability and our study

provides initial evidence that a subset of unclassified missense variants in MLH1 affect these

processes. Another mechanism by which variants may potentially contribute to altered mRNA

expression is through abnormal splicing, and several missense mutations in MLH1 that result in

splicing abnormalities have been reported. Further studies focused on determining the

mechanism by which the MLH1 transcript is regulated will help understand if particular motifs

are associated with transcript regulation and stability. Identification of such motifs will allow for

the more streamlined classification of variants based on their location.

It is possible that the MLH1 expression profile in lymphocytes differs from colon cells. Colon

tissue, if available, would be the ideal source for RNA based analysis. However, unlike

lymphocytes that are readily available from both symptomatic and asymptomatic individuals,

colon tissue is often not easily obtained. Thus, lymphocytes are a useful tool especially for

predictive genetic testing. Furthermore, in current diagnostic practice RT-PCR analysis of RNA

extracted from lymphocytes is used to determine the putative effect of splicing alterations

underscoring its applicability for interpreting the functional implications of genetic alterations.

However, given differences in tissue specific expression, these results should be interpreted with

caution until data comparing the expression profile of MLH1 in colon cells and lymphocytes are

available.

Page 192: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

175

In addition, the PeakPicker methodology that is described in this thesis has the potential to be

applied in the diagnostic setting for the routine assessment of unclassified genetic variants, both

in the MMR system as well as in other genes. The PeakPicker strategy can also be used to further

analyze cases where no genetic lesion has been identified through conventional methodologies.

Even among our panel, two of the variants assessed did not demonstrate allelic imbalances.

Thus, it is likely that a significant proportion of unclassified variants will not demonstrate allelic

imbalances, highlighting the need to use PeakPicker in conjunction with a panel of

functional/biochemical assays. Many of the current functional assessment methods, including the

DNA stability and binding assays described above require molecular cloning, bacterial and cell

culture limiting their applicability in the diagnostic setting. Recently a quantifiable cell-free

MMR activity that can be used for the diagnostic assessment of MLH1 variants was described

[434]. However, this assay focuses on the post-replicative MMR function. Usage of such novel

assays, in conjunction with PeakPicker and standard diagnostic methods such as MSI analysis

and IHC will allow unclassified variants to be characterized more accurately. This also raises

another consideration that is important for this field of study, in that it is unlikely that a single

method will provide a clear cut picture of the pathogenicity associated with an unclassified

genetic variant. Thus, an ideal situation would be to compile a systematic panel of assays, which

can be either extended or compacted based on the clinical and familial information that is

available.

5.5 Functional mechanisms underlying -93G>A variant and its implications

for carcinogenesis

The work presented in this thesis also demonstrates that the MLH1 c.-93G>A promoter

polymorphism alters the transcriptional activity of MLH1. A previous genetic epidemiology

Page 193: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

176

study carried out in our laboratory has shown that the MLH1 -93G>A promoter variant is

strongly associated with MSI tumours [446], a finding that has since been replicated in several

independent populations. Furthermore, -93G>A has been shown to be associated with an

increased risk of endometrial cancer [450]. With the core promoter constructs, we observed

higher activity with the -93G allele compared to the -93A allele in HCT116 colorectal cancer

cells. With the shorter promoter constructs, we consistently observed higher level of activity with

the -93G allele in all cell lines examined. Our findings suggest that this polymorphism has a

subtle but significant and consistent effect at the level of the minimum D promoter region, a

finding that is in line with current epidemiological data. A drastic and universal effect on the

transcription of MLH1 at the level of the core promoter would likely have a larger impact on the

associated cancer risk, which is not consonant with the finding that about 23% of the general,

unaffected, population are carriers of this variant [446]. Less than 5% of all colorectal cancers

are associated with dominant inheritance, while some type of genetic predisposition accounts for

10-25% of all colorectal cancers. Most familial risk is probably attributable in some degree to

inherited low-penetrance alleles that act in combination with environmental risk factors, such as

dietary carcinogens. This is interesting given that the -93 G>A variant has been linked to lifestyle

factors such as smoking [471]. Thus, the finding that the -93G>A variant can alter transcript

levels of MLH1 has important implications for understanding mechanisms at play in moderate-

to low- risk individuals. A previous study found this variant to be associated with MLH1

methylation in endometrial and colorectal tumours [464, 465]. Thus, it is possible that this

variant also acts by altering methylation levels. Our findings also strengthen the genetic

epidemiological findings regarding the association of this variant with endometrial cancer. In

view of this, it may be possible to use MLH1 -93G>A in conjunction with a panel of other

relevant polymorphisms, as prognostic indicators of endometrial cancer risk.

Page 194: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

177

Our studies also indicate that nuclear proteins bind the region of the MLH1 promoter surrounding

the -93G>A variant. In addition, we observe that the -93G>A variant alters the affinity of these

protein interactions. However, these effects are subtle, as would be expected for a common

genetic variant and these findings were replicated in a recent study that looked at the effect of -

93G>A on protein binding in the choriocarcinoma cell line, JEG3 [472]. Despite several

attempts, the identity of the putative factors that bind this region remains to be elucidated. Our

supershift experiments indicate that DEC2 and Max are unlikely to play a significant role in this

process, at least under non-hypoxic conditions in the cell lines examined. A previous study was

able to demonstrate that the MSH6 promoter is regulated by the Sp transcription factors [473]. A

similar approach can be utilized to uncover the identity of the factors that bind the MLH1

promoter region. The identities of these factors will provide clues regarding the mechanisms

regulating MLH1 expression. Moreover, given the different binding patterns we observed

between the colorectal and endometrial cell line, it is possible that considerable variation exists

in the nuclear proteins that bind this region. Accordingly, this should be taken into consideration

when designing experiments that aim to address this question.

This is also the first study to demonstrate that a genetic alteration simultaneously affects both the

regulation of MLH1 and the adjacent gene, EPMAIP1. The cellular role(s) of EPM2AIP1 is

currently not known; although a recent genome-wide association study identified EPM2AIP1 as

a high-priority candidate that modifies breast cancer risk in BRCA2 mutation carriers [457].

EPM2AIP1 was also found to interact with Laforin, a protein implicated in the Wnt signaling

pathway [458]. A recent study reported that deletions in TACSTD1 (the gene directly upstream of

MSH2) led to the somatic silencing of the corresponding MSH2 allele by hypermethylation

[375]. Similarly, it is possible that genetic alterations in EPM2AIP1 can potentially affect MLH1

expression and activity. Alternatively, it is possible that EPM2AIP1 plays a role in

Page 195: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

178

carcinogenesis. This is particularly interesting given that MSI-H tumours show a significant

reduction in MLH1/EPM2AIP1 expression compared with normal colonic mucosa and

microsatellite stable (MSS) tumours [445]. Although methylation of the shared promoter region

is likely to be responsible for this loss of expression in the majority of cases, it is important to

address if EPM2AIP1 independently contributes to neoplasia [342]. Initial studies to address this

should attempt to assess EPM2AIP1 expression by IHC in a subset of MSI-H tumours that show

intact expression of the other known MMR genes. Furthermore, EPM2AIP1‟s role in Lynch

syndrome can be evaluated by sequencing the promoter and coding region of EPM2AIP1 in

individuals with no other known MMR mutations. A knock out model of Epm2aip1 has been

developed by one of our collaborators, Dr. Berge Minassian and his group at the University of

Toronto. It would be of interest to observe if these mice develop tumours, and if so characterize

their phenotype. It is also plausible that EPM2AIP1 does not act independently but cooperates

with MLH1 in tumourigenesis and future studies should address this possibility as well.

Another important theme that arises in this thesis is the classification of genetic alterations. All

the variants assessed in this study are missense substitutions, and with the exception of Q510X

they do not lead to the premature truncation of the protein. However, these genetic variants span

the spectrum in terms of their association with disease risk. For instance, some are the only

genetic alteration identified in kindreds that meet the stringent criteria for Lynch syndrome,

others occur in individuals with suspected but unconfirmed Lynch syndrome, and yet others

occur in individuals unaffected by cancer. They also vary in frequency in that some are novel,

others while rare have been reported in families from different parts of the world, and yet others

occur in a large proportion of the population. With regard to these differences, T117M is

classified as mutation, I19F, R265C, K618A, L749P as unclassified variants and -93G>A as a

single nucleotide polymorphism (SNP). SNPs have traditionally been defined as phenotypically

Page 196: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

179

neutral sequence alterations that are present in the general population with a frequency of >1%

[474]. While lack of involvement in disease is likely true for the large majority of SNPs, various

studies support the notion that a subset of SNPs can potentially contribute to disease

pathogenesis. In fact characterizing unclassified variants in the context of predictive genetic

testing is challenging as it difficult to interpret if they are disease causing mutations responsible

for the phenotype observed or rare, benign polymorphic variants. Our studies demonstrate that at

least with regard to cancer, genetic alterations cannot always be neatly compartmentalized into

defined categories as their consequences form an overlapping continuum. Even with regard to a

single gene such as MLH1, it is likely that the distribution of genetic alterations on this

continuum depend on the complex interactions that occur between other genes and

environmental factors.

MLH1 is an integral component of the MMR system and MMR proteins play a central role in

maintain the stability of the genome. However, MLH1 deficiency and for that matter MMR

deficiency leads to tumours only in specific tissues. This could occur if the threshold of MLH1

required for the diverse functions of the MMR proteins vary between tissues. It could also result

due to the ability of other DNA repair systems to substitute for defective MMR in specific tissues

and the overlap between the functional capabilities of the multiple DNA repair pathways.

Another possibility is that a specific type of error or damage may be more common in certain

tissues, which may in turn influence that the type of repair critical for the integrity of that tissue.

Thus, a combination of these factors may influence the tumour spectrum that arises due to MLH1

deficiency.

In conclusion, the findings presented in this thesis have important implications for the

assessment and study of rare variants identified in suspected Lynch syndrome cases. Our

Page 197: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

180

findings also shed light on the functional mechanisms underlying a common susceptibility

variant. These studies also provide several insights into the regulation and expression of MLH1,

which have implications for furthering our understanding of the MMR system.

The discovery of a predisposing mutation in Lynch syndrome families and the concomitant

increase in surveillance has been shown to significantly reduce mortality [251, 431]. Therefore it

is of paramount importance to classify the pathogenicity associated with unclassified variants to

determine their role in cancer predisposition. Overall, an accurate assessment of the pathogenic

nature of unclassified variants in the MMR genes will greatly increase the accuracy of molecular

diagnostic testing and facilitate more efficient surveillance and prevention of associated cancers.

Page 198: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

181

References

1. Gervaz P, Bucher P, Morel P. Two colons-two cancers: paradigm shift and clinical

implications. J Surg Oncol 2004;88(4):261-6.

2. Thomas LA, Veysey MJ, French G, Hylemon PB, Murphy GM, Dowling RH. Bile acid

metabolism by fresh human colonic contents: a comparison of caecal versus faecal samples. Gut

2001;49(6):835-42.

3. Potten CS, Booth C, Pritchard DM. The intestinal epithelial stem cell: the mucosal

governor. Int J Exp Pathol 1997;78(4):219-43.

4. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell

1990;61(5):759-67.

5. Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, Fan RS, Zborowska

E, Kinzler KW, Vogelstein B, et al. Inactivation of the type II TGF-beta receptor in colon cancer

cells with microsatellite instability. Science 1995;268(5215):1336-8.

6. Thiagalingam S, Lengauer C, Leach FS, Schutte M, Hahn SA, Overhauser J, Willson JK,

Markowitz S, Hamilton SR, Kern SE, et al. Evaluation of candidate tumour suppressor genes on

chromosome 18 in colorectal cancers. Nat Genet 1996;13(3):343-6.

7. Baker SJ, Fearon ER, Nigro JM, Hamilton SR, Preisinger AC, Jessup JM, vanTuinen P,

Ledbetter DH, Barker DF, Nakamura Y, et al. Chromosome 17 deletions and p53 gene mutations

in colorectal carcinomas. Science 1989;244(4901):217-21.

8. Samuels Y, Velculescu VE. Oncogenic mutations of PIK3CA in human cancers. Cell

Cycle 2004;3(10):1221-4.

9. Pino MS, Chung DC. The chromosomal instability pathway in colon cancer.

Gastroenterology 2010;138(6):2059-72.

10. Pretlow TP, Barrow BJ, Ashton WS, O'Riordan MA, Pretlow TG, Jurcisek JA, Stellato

TA. Aberrant crypts: putative preneoplastic foci in human colonic mucosa. Cancer Res

1991;51(5):1564-7.

11. Gupta AK, Pretlow TP, Schoen RE. Aberrant crypt foci: what we know and what we

need to know. Clin Gastroenterol Hepatol 2007;5(5):526-33.

12. Takayama T, Katsuki S, Takahashi Y, Ohi M, Nojiri S, Sakamaki S, Kato J, Kogawa K,

Miyake H, Niitsu Y. Aberrant crypt foci of the colon as precursors of adenoma and cancer. N

Engl J Med 1998;339(18):1277-84.

13. Rudolph RE, Dominitz JA, Lampe JW, Levy L, Qu P, Li SS, Lampe PD, Bronner MP,

Potter JD. Risk factors for colorectal cancer in relation to number and size of aberrant crypt foci

in humans. Cancer Epidemiol Biomarkers Prev 2005;14(3):605-8.

Page 199: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

182

14. Smith AJ, Stern HS, Penner M, Hay K, Mitri A, Bapat BV, Gallinger S. Somatic APC

and K-ras codon 12 mutations in aberrant crypt foci from human colons. Cancer Res

1994;54(21):5527-30.

15. Corpet DE, Tache S. Most effective colon cancer chemopreventive agents in rats: a

systematic review of aberrant crypt foci and tumor data, ranked by potency. Nutr Cancer

2002;43(1):1-21.

16. Gupta AK, Schoen RE. Aberrant crypt foci: are they intermediate endpoints of colon

carcinogenesis in humans? Curr Opin Gastroenterol 2009;25(1):59-65.

17. Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. Cancer statistics, 2007. CA

Cancer J Clin 2007;57(1):43-66.

18. Winawer S, Fletcher R, Rex D, Bond J, Burt R, Ferrucci J, Ganiats T, Levin T, Woolf S,

Johnson D, et al. Colorectal cancer screening and surveillance: clinical guidelines and rationale-

Update based on new evidence. Gastroenterology 2003;124(2):544-60.

19. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J

Clin 2009;59(4):225-49.

20. Canadian Cancer Society's Steering Committee: Canadian Cancer Statistics 2010.

Toronto: Canadian Cancer Society 2010.

21. Canadian Cancer Society's Steering Committee: Canadian Cancer Statistics 2009.

Toronto: Canadian Cancer Society 2009.

22. Whitlock EP, Lin JS, Liles E, Beil TL, Fu R. Screening for colorectal cancer: a targeted,

updated systematic review for the U.S. Preventive Services Task Force. Ann Intern Med

2008;149(9):638-58.

23. Levin B, Lieberman DA, McFarland B, Andrews KS, Brooks D, Bond J, Dash C,

Giardiello FM, Glick S, Johnson D, et al. Screening and surveillance for the early detection of

colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer

Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of

Radiology. Gastroenterology 2008;134(5):1570-95.

24. Winawer SJ, Zauber AG, Ho MN, O'Brien MJ, Gottlieb LS, Sternberg SS, Waye JD,

Schapiro M, Bond JH, Panish JF, et al. Prevention of colorectal cancer by colonoscopic

polypectomy. The National Polyp Study Workgroup. N Engl J Med 1993;329(27):1977-81.

25. Lieberman DA. Clinical practice. Screening for colorectal cancer. N Engl J Med

2009;361(12):1179-87.

26. Hundt S, Haug U, Brenner H. Comparative evaluation of immunochemical fecal occult

blood tests for colorectal adenoma detection. Ann Intern Med 2009;150(3):162-9.

Page 200: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

183

27. Mandel JS, Bond JH, Church TR, Snover DC, Bradley GM, Schuman LM, Ederer F.

Reducing mortality from colorectal cancer by screening for fecal occult blood. Minnesota Colon

Cancer Control Study. N Engl J Med 1993;328(19):1365-71.

28. Hardcastle JD, Chamberlain JO, Robinson MH, Moss SM, Amar SS, Balfour TW, James

PD, Mangham CM. Randomised controlled trial of faecal-occult-blood screening for colorectal

cancer. Lancet 1996;348(9040):1472-7.

29. Kronborg O, Fenger C, Olsen J, Jorgensen OD, Sondergaard O. Randomised study of

screening for colorectal cancer with faecal-occult-blood test. Lancet 1996;348(9040):1467-71.

30. Winawer SJ, Stewart ET, Zauber AG, Bond JH, Ansel H, Waye JD, Hall D, Hamlin JA,

Schapiro M, O'Brien MJ, et al. A comparison of colonoscopy and double-contrast barium enema

for surveillance after polypectomy. National Polyp Study Work Group. N Engl J Med

2000;342(24):1766-72.

31. Lieberman DA, Weiss DG, Bond JH, Ahnen DJ, Garewal H, Chejfec G. Use of

colonoscopy to screen asymptomatic adults for colorectal cancer. Veterans Affairs Cooperative

Study Group 380. N Engl J Med 2000;343(3):162-8.

32. Imperiale TF, Wagner DR, Lin CY, Larkin GN, Rogge JD, Ransohoff DF. Risk of

advanced proximal neoplasms in asymptomatic adults according to the distal colorectal findings.

N Engl J Med 2000;343(3):169-74.

33. Rockey DC, Paulson E, Niedzwiecki D, Davis W, Bosworth HB, Sanders L, Yee J,

Henderson J, Hatten P, Burdick S, et al. Analysis of air contrast barium enema, computed

tomographic colonography, and colonoscopy: prospective comparison. Lancet

2005;365(9456):305-11.

34. Cotton PB, Durkalski VL, Pineau BC, Palesch YY, Mauldin PD, Hoffman B, Vining DJ,

Small WC, Affronti J, Rex D, et al. Computed tomographic colonography (virtual colonoscopy):

a multicenter comparison with standard colonoscopy for detection of colorectal neoplasia. Jama

2004;291(14):1713-9.

35. Soetikno RM, Kaltenbach T, Rouse RV, Park W, Maheshwari A, Sato T, Matsui S,

Friedland S. Prevalence of nonpolypoid (flat and depressed) colorectal neoplasms in

asymptomatic and symptomatic adults. Jama 2008;299(9):1027-35.

36. Leddin D, Hunt R, Champion M, Cockeram A, Flook N, Gould M, Kim YI, Love J,

Morgan D, Natsheh S, et al. Canadian Association of Gastroenterology and the Canadian

Digestive Health Foundation: Guidelines on colon cancer screening. Can J Gastroenterol

2004;18(2):93-9.

37. Imperiale TF, Ransohoff DF, Itzkowitz SH, Turnbull BA, Ross ME. Fecal DNA versus

fecal occult blood for colorectal-cancer screening in an average-risk population. N Engl J Med

2004;351(26):2704-14.

Page 201: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

184

38. Ahlquist DA, Sargent DJ, Loprinzi CL, Levin TR, Rex DK, Ahnen DJ, Knigge K, Lance

MP, Burgart LJ, Hamilton SR, et al. Stool DNA and occult blood testing for screen detection of

colorectal neoplasia. Ann Intern Med 2008;149(7):441-50, W81.

39. Allison JE, Sakoda LC, Levin TR, Tucker JP, Tekawa IS, Cuff T, Pauly MP, Shlager L,

Palitz AM, Zhao WK, et al. Screening for colorectal neoplasms with new fecal occult blood

tests: update on performance characteristics. J Natl Cancer Inst 2007;99(19):1462-70.

40. Itzkowitz SH, Jandorf L, Brand R, Rabeneck L, Schroy PC, 3rd, Sontag S, Johnson D,

Skoletsky J, Durkee K, Markowitz S, et al. Improved fecal DNA test for colorectal cancer

screening. Clin Gastroenterol Hepatol 2007;5(1):111-7.

41. Johnson CD, Chen MH, Toledano AY, Heiken JP, Dachman A, Kuo MD, Menias CO,

Siewert B, Cheema JI, Obregon RG, et al. Accuracy of CT colonography for detection of large

adenomas and cancers. N Engl J Med 2008;359(12):1207-17.

42. Selby JV, Friedman GD, Quesenberry CP, Jr., Weiss NS. A case-control study of

screening sigmoidoscopy and mortality from colorectal cancer. N Engl J Med 1992;326(10):653-

7.

43. Lieberman DA, Holub J, Eisen G, Kraemer D, Morris CD. Prevalence of polyps greater

than 9 mm in a consortium of diverse clinical practice settings in the United States. Clin

Gastroenterol Hepatol 2005;3(8):798-805.

44. Schoenfeld P, Cash B, Flood A, Dobhan R, Eastone J, Coyle W, Kikendall JW, Kim HM,

Weiss DG, Emory T, et al. Colonoscopic screening of average-risk women for colorectal

neoplasia. N Engl J Med 2005;352(20):2061-8.

45. Pickhardt PJ, Choi JR, Hwang I, Butler JA, Puckett ML, Hildebrandt HA, Wong RK,

Nugent PA, Mysliwiec PA, Schindler WR. Computed tomographic virtual colonoscopy to screen

for colorectal neoplasia in asymptomatic adults. N Engl J Med 2003;349(23):2191-200.

46. Singh H, Turner D, Xue L, Targownik LE, Bernstein CN. Risk of developing colorectal

cancer following a negative colonoscopy examination: evidence for a 10-year interval between

colonoscopies. Jama 2006;295(20):2366-73.

47. Baxter NN, Goldwasser MA, Paszat LF, Saskin R, Urbach DR, Rabeneck L. Association

of colonoscopy and death from colorectal cancer. Ann Intern Med 2009;150(1):1-8.

48. Sobin LH, Hermanek P, Hutter RV. TNM classification of malignant tumors. A

comparison between the new (1987) and the old editions. Cancer 1988;61(11):2310-4.

49. Compton CC, Greene FL. The staging of colorectal cancer: 2004 and beyond. CA Cancer

J Clin 2004;54(6):295-308.

50. SEER cancer statistics review 2000. 2003.

51. Aslam MI, Kelkar A, Sharpe D, Jameson JS. Ten years experience of managing the

primary tumours in patients with stage IV colorectal cancers. Int J Surg.

Page 202: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

185

52. Derwinger K, Kodeda K, Bexe-Lindskog E, Taflin H. Tumour differentiation grade is

associated with TNM staging and the risk of node metastasis in colorectal cancer. Acta

Oncol;49(1):57-62.

53. Green FL, Page DL, Fleming ID, Fritz A, Balch CM, Haller DG, Morrow M. AJCC

Cancer Staging Manual, 6th Edition New York, NY: Springer; 2002.

54. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100(1):57-70.

55. Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, Mandelker D, Leary RJ,

Ptak J, Silliman N, et al. The consensus coding sequences of human breast and colorectal

cancers. Science 2006;314(5797):268-74.

56. Albertini RJ, Nicklas JA, O'Neill JP, Robison SH. In vivo somatic mutations in humans:

measurement and analysis. Annu Rev Genet 1990;24:305-26.

57. Loeb LA. Mutator phenotype may be required for multistage carcinogenesis. Cancer Res

1991;51(12):3075-9.

58. Loeb LA. A mutator phenotype in cancer. Cancer Res 2001;61(8):3230-9.

59. Sinicrope FA, Rego RL, Halling KC, Foster N, Sargent DJ, La Plant B, French AJ,

Laurie JA, Goldberg RM, Thibodeau SN, et al. Prognostic impact of microsatellite instability

and DNA ploidy in human colon carcinoma patients. Gastroenterology 2006;131(3):729-37.

60. Shen L, Toyota M, Kondo Y, Lin E, Zhang L, Guo Y, Hernandez NS, Chen X, Ahmed S,

Konishi K, et al. Integrated genetic and epigenetic analysis identifies three different subclasses of

colon cancer. Proc Natl Acad Sci U S A 2007;104(47):18654-9.

61. Cheng YW, Pincas H, Bacolod MD, Schemmann G, Giardina SF, Huang J, Barral S,

Idrees K, Khan SA, Zeng Z, et al. CpG island methylator phenotype associates with low-degree

chromosomal abnormalities in colorectal cancer. Clin Cancer Res 2008;14(19):6005-13.

62. Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature

1998;396(6712):643-9.

63. Ogino S, Goel A. Molecular classification and correlates in colorectal cancer. J Mol

Diagn 2008;10(1):13-27.

64. Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer

pathogenesis. Gastroenterology 2008;135(4):1079-99.

65. Narayan S, Roy D. Role of APC and DNA mismatch repair genes in the development of

colorectal cancers. Mol Cancer 2003;2:41.

66. Rubinfeld B, Souza B, Albert I, Muller O, Chamberlain SH, Masiarz FR, Munemitsu S,

Polakis P. Association of the APC gene product with beta-catenin. Science

1993;262(5140):1731-4.

Page 203: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

186

67. Su LK, Vogelstein B, Kinzler KW. Association of the APC tumor suppressor protein

with catenins. Science 1993;262(5140):1734-7.

68. Rubinfeld B, Robbins P, El-Gamil M, Albert I, Porfiri E, Polakis P. Stabilization of beta-

catenin by genetic defects in melanoma cell lines. Science 1997;275(5307):1790-2.

69. He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein

B, Kinzler KW. Identification of c-MYC as a target of the APC pathway. Science

1998;281(5382):1509-12.

70. Muhua L, Adames NR, Murphy MD, Shields CR, Cooper JA. A cytokinesis checkpoint

requiring the yeast homologue of an APC-binding protein. Nature 1998;393(6684):487-91.

71. Fodde R, Kuipers J, Rosenberg C, Smits R, Kielman M, Gaspar C, van Es JH, Breukel C,

Wiegant J, Giles RH, et al. Mutations in the APC tumour suppressor gene cause chromosomal

instability. Nat Cell Biol 2001;3(4):433-8.

72. Sieber OM, Heinimann K, Tomlinson IP. Genomic instability--the engine of

tumorigenesis? Nat Rev Cancer 2003;3(9):701-8.

73. Jallepalli PV, Lengauer C. Chromosome segregation and cancer: cutting through the

mystery. Nat Rev Cancer 2001;1(2):109-17.

74. Boveri T. Zur Frage der Enstehung Maligner Tumoren 1914.

75. von Hansemann D. Virchows Arch. Pathol. Anat 1890;119:299-326.

76. Jefford CE, Irminger-Finger I. Mechanisms of chromosome instability in cancers. Crit

Rev Oncol Hematol 2006;59(1):1-14.

77. Cahill DP, Lengauer C, Yu J, Riggins GJ, Willson JK, Markowitz SD, Kinzler KW,

Vogelstein B. Mutations of mitotic checkpoint genes in human cancers. Nature

1998;392(6673):300-3.

78. Yu H, Peters JM, King RW, Page AM, Hieter P, Kirschner MW. Identification of a cullin

homology region in a subunit of the anaphase-promoting complex. Science

1998;279(5354):1219-22.

79. Harper JW. A phosphorylation-driven ubiquitination switch for cell-cycle control. Trends

Cell Biol 2002;12(3):104-7.

80. Peters JM. The anaphase-promoting complex: proteolysis in mitosis and beyond. Mol

Cell 2002;9(5):931-43.

81. Nasmyth K. Segregating sister genomes: the molecular biology of chromosome

separation. Science 2002;297(5581):559-65.

82. Nasmyth K, Peters JM, Uhlmann F. Splitting the chromosome: cutting the ties that bind

sister chromatids. Science 2000;288(5470):1379-85.

Page 204: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

187

83. Jallepalli PV, Waizenegger IC, Bunz F, Langer S, Speicher MR, Peters JM, Kinzler KW,

Vogelstein B, Lengauer C. Securin is required for chromosomal stability in human cells. Cell

2001;105(4):445-57.

84. Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in colorectal cancers. Nature

1997;386(6625):623-7.

85. Yuen KW, Warren CD, Chen O, Kwok T, Hieter P, Spencer FA. Systematic genome

instability screens in yeast and their potential relevance to cancer. Proc Natl Acad Sci U S A

2007;104(10):3925-30.

86. Brinkley BR. Managing the centrosome numbers game: from chaos to stability in cancer

cell division. Trends Cell Biol 2001;11(1):18-21.

87. D'Assoro AB, Lingle WL, Salisbury JL. Centrosome amplification and the development

of cancer. Oncogene 2002;21(40):6146-53.

88. Zhou H, Kuang J, Zhong L, Kuo WL, Gray JW, Sahin A, Brinkley BR, Sen S. Tumour

amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and

transformation. Nat Genet 1998;20(2):189-93.

89. Bischoff JR, Anderson L, Zhu Y, Mossie K, Ng L, Souza B, Schryver B, Flanagan P,

Clairvoyant F, Ginther C, et al. A homologue of Drosophila aurora kinase is oncogenic and

amplified in human colorectal cancers. Embo J 1998;17(11):3052-65.

90. Takahashi T, Sano B, Nagata T, Kato H, Sugiyama Y, Kunieda K, Kimura M, Okano Y,

Saji S. Polo-like kinase 1 (PLK1) is overexpressed in primary colorectal cancers. Cancer Sci

2003;94(2):148-52.

91. Basto R, Brunk K, Vinadogrova T, Peel N, Franz A, Khodjakov A, Raff JW. Centrosome

amplification can initiate tumorigenesis in flies. Cell 2008;133(6):1032-42.

92. Hoeijmakers JH. Genome maintenance mechanisms for preventing cancer. Nature

2001;411(6835):366-74.

93. Counter CM, Avilion AA, LeFeuvre CE, Stewart NG, Greider CW, Harley CB, Bacchetti

S. Telomere shortening associated with chromosome instability is arrested in immortal cells

which express telomerase activity. Embo J 1992;11(5):1921-9.

94. Harley CB. Telomeres and Aging. In: Blackburn EHaG, C.W., editor. Telomeres. Cold

Spring Harbour: Cold Spring Harbour Press; 1995.

95. Gonzalez-Suarez E, Samper E, Flores JM, Blasco MA. Telomerase-deficient mice with

short telomeres are resistant to skin tumorigenesis. Nat Genet 2000;26(1):114-7.

96. Greenberg RA, Chin L, Femino A, Lee KH, Gottlieb GJ, Singer RH, Greider CW,

DePinho RA. Short dysfunctional telomeres impair tumorigenesis in the INK4a(delta2/3) cancer-

prone mouse. Cell 1999;97(4):515-25.

Page 205: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

188

97. Rudolph KL, Chang S, Lee HW, Blasco M, Gottlieb GJ, Greider C, DePinho RA.

Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 1999;96(5):701-

12.

98. Rudolph KL, Millard M, Bosenberg MW, DePinho RA. Telomere dysfunction and

evolution of intestinal carcinoma in mice and humans. Nat Genet 2001;28(2):155-9.

99. Chadeneau C, Hay K, Hirte HW, Gallinger S, Bacchetti S. Telomerase activity associated

with acquisition of malignancy in human colorectal cancer. Cancer Res 1995;55(12):2533-6.

100. Engelhardt M, Drullinsky P, Guillem J, Moore MA. Telomerase and telomere length in

the development and progression of premalignant lesions to colorectal cancer. Clin Cancer Res

1997;3(11):1931-41.

101. Tatsumoto N, Hiyama E, Murakami Y, Imamura Y, Shay JW, Matsuura Y, Yokoyama T.

High telomerase activity is an independent prognostic indicator of poor outcome in colorectal

cancer. Clin Cancer Res 2000;6(7):2696-701.

102. Reddel RR. Alternative lengthening of telomeres, telomerase, and cancer. Cancer Lett

2003;194(2):155-62.

103. de Lange T. Telomere dynamics and genome instability in human cancer. In: Blackburn

EHaG, C.W., editor. Telomeres. Cold Spring Harbour: Cold Spring Harbour Press; 1995. p. 265-

293.

104. Nakamura K, Furugori E, Esaki Y, Arai T, Sawabe M, Okayasu I, Fujiwara M, Kammori

M, Mafune K, Kato M, et al. Correlation of telomere lengths in normal and cancers tissue in the

large bowel. Cancer Lett 2000;158(2):179-84.

105. Takagi S, Kinouchi Y, Hiwatashi N, Chida M, Nagashima F, Takahashi S, Negoro K,

Shimosegawa T, Toyota T. Telomere shortening and the clinicopathologic characteristics of

human colorectal carcinomas. Cancer 1999;86(8):1431-6.

106. Gertler R, Rosenberg R, Stricker D, Friederichs J, Hoos A, Werner M, Ulm K, Holzmann

B, Nekarda H, Siewert JR. Telomere length and human telomerase reverse transcriptase

expression as markers for progression and prognosis of colorectal carcinoma. J Clin Oncol

2004;22(10):1807-14.

107. Meeker AK, Hicks JL, Iacobuzio-Donahue CA, Montgomery EA, Westra WH, Chan TY,

Ronnett BM, De Marzo AM. Telomere length abnormalities occur early in the initiation of

epithelial carcinogenesis. Clin Cancer Res 2004;10(10):3317-26.

108. Nyberg KA, Michelson RJ, Putnam CW, Weinert TA. Toward maintaining the genome:

DNA damage and replication checkpoints. Annu Rev Genet 2002;36:617-56.

109. Bao S, Chang MS, Auclair D, Sun Y, Wang Y, Wong WK, Zhang J, Liu Y, Qian X,

Sutherland R, et al. HRad17, a human homologue of the Schizosaccharomyces pombe

checkpoint gene rad17, is overexpressed in colon carcinoma. Cancer Res 1999;59(9):2023-8.

Page 206: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

189

110. Rotman G, Shiloh Y. ATM: from gene to function. Hum Mol Genet 1998;7(10):1555-63.

111. Smith L, Liu SJ, Goodrich L, Jacobson D, Degnin C, Bentley N, Carr A, Flaggs G,

Keegan K, Hoekstra M, et al. Duplication of ATR inhibits MyoD, induces aneuploidy and

eliminates radiation-induced G1 arrest. Nat Genet 1998;19(1):39-46.

112. Zhang H, Tombline G, Weber BL. BRCA1, BRCA2, and DNA damage response:

collision or collusion? Cell 1998;92(4):433-6.

113. Liu G, Parant JM, Lang G, Chau P, Chavez-Reyes A, El-Naggar AK, Multani A, Chang

S, Lozano G. Chromosome stability, in the absence of apoptosis, is critical for suppression of

tumorigenesis in Trp53 mutant mice. Nat Genet 2004;36(1):63-8.

114. Kolodner RD, Putnam CD, Myung K. Maintenance of genome stability in

Saccharomyces cerevisiae. Science 2002;297(5581):552-7.

115. Wood RD, Mitchell M, Sgouros J, Lindahl T. Human DNA repair genes. Science

2001;291(5507):1284-9.

116. Barnes DE, Lindahl T, Sedgwick B. DNA repair. Curr Opin Cell Biol 1993;5(3):424-33.

117. Chan KK, Zhang QM, Dianov GL. Base excision repair fidelity in normal and cancer

cells. Mutagenesis 2006;21(3):173-8.

118. Al-Tassan N, Chmiel NH, Maynard J, Fleming N, Livingston AL, Williams GT, Hodges

AK, Davies DR, David SS, Sampson JR, et al. Inherited variants of MYH associated with

somatic G:C-->T:A mutations in colorectal tumors. Nat Genet 2002;30(2):227-32.

119. Cardoso J, Molenaar L, de Menezes RX, van Leerdam M, Rosenberg C, Moslein G,

Sampson J, Morreau H, Boer JM, Fodde R. Chromosomal instability in MYH- and APC-mutant

adenomatous polyps. Cancer Res 2006;66(5):2514-9.

120. Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer

SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN, et al. A National Cancer Institute Workshop on

Microsatellite Instability for cancer detection and familial predisposition: development of

international criteria for the determination of microsatellite instability in colorectal cancer.

Cancer Res 1998;58(22):5248-57.

121. Kane MF, Loda M, Gaida GM, Lipman J, Mishra R, Goldman H, Jessup JM, Kolodner

R. Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic

colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res 1997;57(5):808-

11.

122. Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, Markowitz S, Willson JK,

Hamilton SR, Kinzler KW, et al. Incidence and functional consequences of hMLH1 promoter

hypermethylation in colorectal carcinoma. Proc Natl Acad Sci U S A 1998;95(12):6870-5.

Page 207: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

190

123. Aaltonen LA, Peltomaki P, Leach FS, Sistonen P, Pylkkanen L, Mecklin JP, Jarvinen H,

Powell SM, Jen J, Hamilton SR, et al. Clues to the pathogenesis of familial colorectal cancer.

Science 1993;260(5109):812-6.

124. Aaltonen LA, Peltomaki P, Mecklin JP, Jarvinen H, Jass JR, Green JS, Lynch HT,

Watson P, Tallqvist G, Juhola M, et al. Replication errors in benign and malignant tumors from

hereditary nonpolyposis colorectal cancer patients. Cancer Res 1994;54(7):1645-8.

125. Jenkins MA, Hayashi S, O'Shea AM, Burgart LJ, Smyrk TC, Shimizu D, Waring PM,

Ruszkiewicz AR, Pollett AF, Redston M, et al. Pathology features in Bethesda guidelines predict

colorectal cancer microsatellite instability: a population-based study. Gastroenterology

2007;133(1):48-56.

126. Popat S, Hubner R, Houlston RS. Systematic review of microsatellite instability and

colorectal cancer prognosis. J Clin Oncol 2005;23(3):609-18.

127. Alexander J, Watanabe T, Wu TT, Rashid A, Li S, Hamilton SR. Histopathological

identification of colon cancer with microsatellite instability. Am J Pathol 2001;158(2):527-35.

128. Gryfe R, Kim H, Hsieh ET, Aronson MD, Holowaty EJ, Bull SB, Redston M, Gallinger

S. Tumor microsatellite instability and clinical outcome in young patients with colorectal cancer.

N Engl J Med 2000;342(2):69-77.

129. Woerner SM, Yuan YP, Benner A, Korff S, von Knebel Doeberitz M, Bork P.

SelTarbase, a database of human mononucleotide-microsatellite mutations and their potential

impact to tumorigenesis and immunology. Nucleic Acids Res 2010;38(Database issue):D682-9.

130. Parsons R, Myeroff LL, Liu B, Willson JK, Markowitz SD, Kinzler KW, Vogelstein B.

Microsatellite instability and mutations of the transforming growth factor beta type II receptor

gene in colorectal cancer. Cancer Res 1995;55(23):5548-50.

131. LeRoith D, Roberts CT, Jr. The insulin-like growth factor system and cancer. Cancer Lett

2003;195(2):127-37.

132. Souza RF, Appel R, Yin J, Wang S, Smolinski KN, Abraham JM, Zou TT, Shi YQ, Lei J,

Cottrell J, et al. Microsatellite instability in the insulin-like growth factor II receptor gene in

gastrointestinal tumours. Nat Genet 1996;14(3):255-7.

133. Saeki H, Tanaka S, Tokunaga E, Kawaguchi H, Ikeda Y, Maehara Y, Sugimachi K.

Genetic alterations in the human Tcf-4 gene in Japanese patients with sporadic gastrointestinal

cancers with microsatellite instability. Oncology 2001;61(2):156-61.

134. Duval A, Gayet J, Zhou XP, Iacopetta B, Thomas G, Hamelin R. Frequent frameshift

mutations of the TCF-4 gene in colorectal cancers with microsatellite instability. Cancer Res

1999;59(17):4213-5.

135. Eastman Q, Grosschedl R. Regulation of LEF-1/TCF transcription factors by Wnt and

other signals. Curr Opin Cell Biol 1999;11(2):233-40.

Page 208: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

191

136. Ruckert S, Hiendlmeyer E, Brueckl WM, Oswald U, Beyser K, Dietmaier W, Haynl A,

Koch C, Ruschoff J, Brabletz T, et al. T-cell factor-4 frameshift mutations occur frequently in

human microsatellite instability-high colorectal carcinomas but do not contribute to

carcinogenesis. Cancer Res 2002;62(11):3009-13.

137. Shin KH, Park YJ, Park JG. PTEN gene mutations in colorectal cancers displaying

microsatellite instability. Cancer Lett 2001;174(2):189-94.

138. Guanti G, Resta N, Simone C, Cariola F, Demma I, Fiorente P, Gentile M. Involvement

of PTEN mutations in the genetic pathways of colorectal cancerogenesis. Hum Mol Genet

2000;9(2):283-7.

139. Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC, Perucho M. Somatic

frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype.

Science 1997;275(5302):967-9.

140. Malkhosyan S, Rampino N, Yamamoto H, Perucho M. Frameshift mutator mutations.

Nature 1996;382(6591):499-500.

141. Saetta AA, Korkolopoulou P, Karlou M, Levidou G, Goudopoulou A, Thymara I,

Stamatelli A, Tzivras M, Michalopoulos NV, Thomas-Tsagli E, et al. TGF-betaRII, BAX,

IGFIIR, caspase-5, hMSH3 and hMSH6 alterations are not associated with microsatellite

instability or p53 mutations in invasive urothelial carcinoma of the urinary bladder. Pathology

2007;39(4):425-32.

142. Grady WM, Rajput A, Myeroff L, Liu DF, Kwon K, Willis J, Markowitz S. Mutation of

the type II transforming growth factor-beta receptor is coincident with the transformation of

human colon adenomas to malignant carcinomas. Cancer Res 1998;58(14):3101-4.

143. Yagi OK, Akiyama Y, Nomizu T, Iwama T, Endo M, Yuasa Y. Proapoptotic gene BAX

is frequently mutated in hereditary nonpolyposis colorectal cancers but not in adenomas.

Gastroenterology 1998;114(2):268-74.

144. Vasen HF, Wijnen JT, Menko FH, Kleibeuker JH, Taal BG, Griffioen G, Nagengast FM,

Meijers-Heijboer EH, Bertario L, Varesco L, et al. Cancer risk in families with hereditary

nonpolyposis colorectal cancer diagnosed by mutation analysis. Gastroenterology

1996;110(4):1020-7.

145. Tomlinson I, Halford S, Aaltonen L, Hawkins N, Ward R. Does MSI-low exist? J Pathol

2002;197(1):6-13.

146. Halford S, Sasieni P, Rowan A, Wasan H, Bodmer W, Talbot I, Hawkins N, Ward R,

Tomlinson I. Low-level microsatellite instability occurs in most colorectal cancers and is a

nonrandomly distributed quantitative trait. Cancer Res 2002;62(1):53-7.

147. Jass JR. Classification of colorectal cancer based on correlation of clinical,

morphological and molecular features. Histopathology 2007;50(1):113-30.

Page 209: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

192

148. Mori Y, Selaru FM, Sato F, Yin J, Simms LA, Xu Y, Olaru A, Deacu E, Wang S, Taylor

JM, et al. The impact of microsatellite instability on the molecular phenotype of colorectal

tumors. Cancer Res 2003;63(15):4577-82.

149. Iglesias D, Fernandez-Peralta AM, Nejda N, Daimiel L, Azcoita MM, Oliart S, Gonzalez-

Aguilera JJ. RIS1, a gene with trinucleotide repeats, is a target in the mutator pathway of

colorectal carcinogenesis. Cancer Genet Cytogenet 2006;167(2):138-44.

150. Kohonen-Corish MR, Daniel JJ, Chan C, Lin BP, Kwun SY, Dent OF, Dhillon VS, Trent

RJ, Chapuis PH, Bokey EL. Low microsatellite instability is associated with poor prognosis in

stage C colon cancer. J Clin Oncol 2005;23(10):2318-24.

151. Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton

SR, Laurent-Puig P, Gryfe R, Shepherd LE, et al. Tumor microsatellite-instability status as a

predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J

Med 2003;349(3):247-57.

152. Jover R, Zapater P, Castells A, Llor X, Andreu M, Cubiella J, Balaguer F, Sempere L,

Xicola RM, Bujanda L, et al. The efficacy of adjuvant chemotherapy with 5-fluorouracil in

colorectal cancer depends on the mismatch repair status. Eur J Cancer 2008.

153. Valeri N, Gasparini P, Fabbri M, Braconi C, Veronese A, Lovat F, Adair B, Vannini I,

Fanini F, Bottoni A, et al. Modulation of mismatch repair and genomic stability by miR-155.

Proc Natl Acad Sci U S A 2010;107(15):6982-7.

154. Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, Poon TC, Ng SS, Sung JJ.

Differential expression of microRNAs in plasma of patients with colorectal cancer: a potential

marker for colorectal cancer screening. Gut 2009;58(10):1375-81.

155. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M,

Roldo C, Ferracin M, et al. A microRNA expression signature of human solid tumors defines

cancer gene targets. Proc Natl Acad Sci U S A 2006;103(7):2257-61.

156. Lanza G, Ferracin M, Gafa R, Veronese A, Spizzo R, Pichiorri F, Liu CG, Calin GA,

Croce CM, Negrini M. mRNA/microRNA gene expression profile in microsatellite unstable

colorectal cancer. Mol Cancer 2007;6:54.

157. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K,

Doyle M, FitzHugh W, et al. Initial sequencing and analysis of the human genome. Nature

2001;409(6822):860-921.

158. Jacobsen SE. Gene silencing: Maintaining methylation patterns. Curr Biol

1999;9(16):R617-9.

159. Park Y, Kuroda MI. Epigenetic aspects of X-chromosome dosage compensation. Science

2001;293(5532):1083-5.

160. Feinberg AP, Tycko B. The history of cancer epigenetics. Nat Rev Cancer

2004;4(2):143-53.

Page 210: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

193

161. Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers

from their normal counterparts. Nature 1983;301(5895):89-92.

162. Golbus J, Palella TD, Richardson BC. Quantitative changes in T cell DNA methylation

occur during differentiation and ageing. Eur J Immunol 1990;20(8):1869-72.

163. Richardson B. Impact of aging on DNA methylation. Ageing Res Rev 2003;2(3):245-61.

164. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002;16(1):6-21.

165. Li H, Myeroff L, Smiraglia D, Romero MF, Pretlow TP, Kasturi L, Lutterbaugh J, Rerko

RM, Casey G, Issa JP, et al. SLC5A8, a sodium transporter, is a tumor suppressor gene silenced

by methylation in human colon aberrant crypt foci and cancers. Proc Natl Acad Sci U S A

2003;100(14):8412-7.

166. Moinova HR, Chen WD, Shen L, Smiraglia D, Olechnowicz J, Ravi L, Kasturi L,

Myeroff L, Plass C, Parsons R, et al. HLTF gene silencing in human colon cancer. Proc Natl

Acad Sci U S A 2002;99(7):4562-7.

167. Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island

methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A 1999;96(15):8681-6.

168. Ogino S, Cantor M, Kawasaki T, Brahmandam M, Kirkner GJ, Weisenberger DJ,

Campan M, Laird PW, Loda M, Fuchs CS. CpG island methylator phenotype (CIMP) of

colorectal cancer is best characterised by quantitative DNA methylation analysis and prospective

cohort studies. Gut 2006;55(7):1000-6.

169. Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH,

Widschwendter M, Weener D, Buchanan D, et al. CpG island methylator phenotype underlies

sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal

cancer. Nat Genet 2006;38(7):787-93.

170. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J,

Weissman IL, Wahl GM. Cancer Stem Cells--Perspectives on Current Status and Future

Directions: AACR Workshop on Cancer Stem Cells. Cancer Res 2006;66(19):9339-44.

171. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that

originates from a primitive hematopoietic cell. Nat Med 1997;3(7):730-7.

172. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective

identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100(7):3983-8.

173. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB.

Identification of a cancer stem cell in human brain tumors. Cancer Res 2003;63(18):5821-8.

174. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM,

Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature

2004;432(7015):396-401.

Page 211: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

194

175. O'Brien C A, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of

initiating tumour growth in immunodeficient mice. Nature 2006.

176. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A,

Korving J, Begthel H, Peters PJ, et al. Identification of stem cells in small intestine and colon by

marker gene Lgr5. Nature 2007;449(7165):1003-7.

177. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M,

Danenberg E, Clarke AR, Sansom OJ, Clevers H. Crypt stem cells as the cells-of-origin of

intestinal cancer. Nature 2009;457(7229):608-11.

178. Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de Jong JH, Borovski T,

Tuynman JB, Todaro M, Merz C, Rodermond H, et al. Wnt activity defines colon cancer stem

cells and is regulated by the microenvironment. Nat Cell Biol 2010;12(5):468-76.

179. Slattery ML, Boucher KM, Caan BJ, Potter JD, Ma KN. Eating patterns and risk of colon

cancer. Am J Epidemiol 1998;148(1):4-16.

180. Terry P, Giovannucci E, Michels KB, Bergkvist L, Hansen H, Holmberg L, Wolk A.

Fruit, vegetables, dietary fiber, and risk of colorectal cancer. J Natl Cancer Inst 2001;93(7):525-

33.

181. Sandhu MS, White IR, McPherson K. Systematic review of the prospective cohort

studies on meat consumption and colorectal cancer risk: a meta-analytical approach. Cancer

Epidemiol Biomarkers Prev 2001;10(5):439-46.

182. Lin OS. Acquired risk factors for colorectal cancer. Methods Mol Biol 2009;472:361-72.

183. Ghoshal A, Preisegger KH, Takayama S, Thorgeirsson SS, Snyderwine EG. Induction of

mammary tumors in female Sprague-Dawley rats by the food-derived carcinogen 2-amino-1-

methyl-6-phenylimidazo[4,5-b]pyridine and effect of dietary fat. Carcinogenesis

1994;15(11):2429-33.

184. Layton DW, Bogen KT, Knize MG, Hatch FT, Johnson VM, Felton JS. Cancer risk of

heterocyclic amines in cooked foods: an analysis and implications for research. Carcinogenesis

1995;16(1):39-52.

185. Shirai T, Sano M, Tamano S, Takahashi S, Hirose M, Futakuchi M, Hasegawa R, Imaida

K, Matsumoto K, Wakabayashi K, et al. The prostate: a target for carcinogenicity of 2-amino-1-

methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) derived from cooked foods. Cancer Res

1997;57(2):195-8.

186. Tudek B, Bird RP, Bruce WR. Foci of aberrant crypts in the colons of mice and rats

exposed to carcinogens associated with foods. Cancer Res 1989;49(5):1236-40.

187. Sugimura T, Terada M. Experimental chemical carcinogenesis in the stomach and colon.

Jpn J Clin Oncol 1998;28(3):163-7.

Page 212: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

195

188. Bardelli A, Cahill DP, Lederer G, Speicher MR, Kinzler KW, Vogelstein B, Lengauer C.

Carcinogen-specific induction of genetic instability. Proc Natl Acad Sci U S A

2001;98(10):5770-5.

189. Pedersen A, Johansen C, Gronbaek M. Relations between amount and type of alcohol and

colon and rectal cancer in a Danish population based cohort study. Gut 2003;52(6):861-7.

190. Shimizu N, Nagata C, Shimizu H, Kametani M, Takeyama N, Ohnuma T, Matsushita S.

Height, weight, and alcohol consumption in relation to the risk of colorectal cancer in Japan: a

prospective study. Br J Cancer 2003;88(7):1038-43.

191. Cho E, Smith-Warner SA, Ritz J, van den Brandt PA, Colditz GA, Folsom AR,

Freudenheim JL, Giovannucci E, Goldbohm RA, Graham S, et al. Alcohol intake and colorectal

cancer: a pooled analysis of 8 cohort studies. Ann Intern Med 2004;140(8):603-13.

192. Moskal A, Norat T, Ferrari P, Riboli E. Alcohol intake and colorectal cancer risk: a dose-

response meta-analysis of published cohort studies. Int J Cancer 2006;120(3):664-71.

193. Seitz HK, Stickel F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nat Rev

Cancer 2007;7(8):599-612.

194. Nilsen TI, Vatten LJ. Prospective study of colorectal cancer risk and physical activity,

diabetes, blood glucose and BMI: exploring the hyperinsulinaemia hypothesis. Br J Cancer

2001;84(3):417-22.

195. Botteri E, Iodice S, Bagnardi V, Raimondi S, Lowenfels AB, Maisonneuve P. Smoking

and colorectal cancer: a meta-analysis. JAMA 2008;300(23):2765-78.

196. Ekbom A, Helmick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer. A

population-based study. N Engl J Med 1990;323(18):1228-33.

197. Ekbom A, Helmick C, Zack M, Adami HO. Increased risk of large-bowel cancer in

Crohn's disease with colonic involvement. Lancet 1990;336(8711):357-9.

198. Kornbluth A, Sachar DB. Ulcerative colitis practice guidelines in adults (update):

American College of Gastroenterology, Practice Parameters Committee. Am J Gastroenterol

2004;99(7):1371-85.

199. Rutter M, Saunders B, Wilkinson K, Rumbles S, Schofield G, Kamm M, Williams C,

Price A, Talbot I, Forbes A. Severity of inflammation is a risk factor for colorectal neoplasia in

ulcerative colitis. Gastroenterology 2004;126(2):451-9.

200. Kinzler KW, Nilbert MC, Su LK, Vogelstein B, Bryan TM, Levy DB, Smith KJ,

Preisinger AC, Hedge P, McKechnie D, et al. Identification of FAP locus genes from

chromosome 5q21. Science 1991;253(5020):661-5.

201. Burke C. Risk stratification for periampullary carcinoma in patients with familial

adenomatous polyposis: does theodore know what to do now? Gastroenterology

2001;121(5):1246-8.

Page 213: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

196

202. Park YJ, Shin KH, Park JG. Risk of gastric cancer in hereditary nonpolyposis colorectal

cancer in Korea. Clin Cancer Res 2000;6(8):2994-8.

203. Rowan AJ, Lamlum H, Ilyas M, Wheeler J, Straub J, Papadopoulou A, Bicknell D,

Bodmer WF, Tomlinson IP. APC mutations in sporadic colorectal tumors: A mutational

"hotspot" and interdependence of the "two hits". Proc Natl Acad Sci U S A 2000;97(7):3352-7.

204. Lamlum H, Ilyas M, Rowan A, Clark S, Johnson V, Bell J, Frayling I, Efstathiou J, Pack

K, Payne S, et al. The type of somatic mutation at APC in familial adenomatous polyposis is

determined by the site of the germline mutation: a new facet to Knudson's 'two-hit' hypothesis.

Nat Med 1999;5(9):1071-5.

205. Nagase H, Miyoshi Y, Horii A, Aoki T, Ogawa M, Utsunomiya J, Baba S, Sasazuki T,

Nakamura Y. Correlation between the location of germ-line mutations in the APC gene and the

number of colorectal polyps in familial adenomatous polyposis patients. Cancer Res

1992;52(14):4055-7.

206. Spirio L, Olschwang S, Groden J, Robertson M, Samowitz W, Joslyn G, Gelbert L,

Thliveris A, Carlson M, Otterud B, et al. Alleles of the APC gene: an attenuated form of familial

polyposis. Cell 1993;75(5):951-7.

207. Knudsen AL, Bisgaard ML, Bulow S. Attenuated familial adenomatous polyposis

(AFAP). A review of the literature. Fam Cancer 2003;2(1):43-55.

208. de la Chapelle A. Genetic predisposition to colorectal cancer. Nat Rev Cancer

2004;4(10):769-80.

209. Sampson JR, Dolwani S, Jones S, Eccles D, Ellis A, Evans DG, Frayling I, Jordan S,

Maher ER, Mak T, et al. Autosomal recessive colorectal adenomatous polyposis due to inherited

mutations of MYH. Lancet 2003;362(9377):39-41.

210. Sieber OM, Lipton L, Crabtree M, Heinimann K, Fidalgo P, Phillips RK, Bisgaard ML,

Orntoft TF, Aaltonen LA, Hodgson SV, et al. Multiple colorectal adenomas, classic

adenomatous polyposis, and germ-line mutations in MYH. N Engl J Med 2003;348(9):791-9.

211. Shibutani S, Takeshita M, Grollman AP. Insertion of specific bases during DNA

synthesis past the oxidation-damaged base 8-oxodG. Nature 1991;349(6308):431-4.

212. Croitoru ME, Cleary SP, Di Nicola N, Manno M, Selander T, Aronson M, Redston M,

Cotterchio M, Knight J, Gryfe R, et al. Association between biallelic and monoallelic germline

MYH gene mutations and colorectal cancer risk. J Natl Cancer Inst 2004;96(21):1631-4.

213. Jones S, Emmerson P, Maynard J, Best JM, Jordan S, Williams GT, Sampson JR,

Cheadle JP. Biallelic germline mutations in MYH predispose to multiple colorectal adenoma and

somatic G:C-->T:A mutations. Hum Mol Genet 2002;11(23):2961-7.

214. Schrager CA, Schneider D, Gruener AC, Tsou HC, Peacocke M. Clinical and

pathological features of breast disease in Cowden's syndrome: an underrecognized syndrome

with an increased risk of breast cancer. Hum Pathol 1998;29(1):47-53.

Page 214: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

197

215. Starink TM, van der Veen JP, Arwert F, de Waal LP, de Lange GG, Gille JJ, Eriksson

AW. The Cowden syndrome: a clinical and genetic study in 21 patients. Clin Genet

1986;29(3):222-33.

216. Gammon A, Jasperson K, Kohlmann W, Burt RW. Hamartomatous polyposis syndromes.

Best Pract Res Clin Gastroenterol 2009;23(2):219-31.

217. Salem OS, Steck WD. Cowden's disease (multiple hamartoma and neoplasia syndrome).

A case report and review of the English literature. J Am Acad Dermatol 1983;8(5):686-96.

218. Eng C. Constipation, polyps, or cancer? Let PTEN predict your future. Am J Med Genet

A 2003;122A(4):315-22.

219. Amos CI, Keitheri-Cheteri MB, Sabripour M, Wei C, McGarrity TJ, Seldin MF, Nations

L, Lynch PM, Fidder HH, Friedman E, et al. Genotype-phenotype correlations in Peutz-Jeghers

syndrome. J Med Genet 2004;41(5):327-33.

220. Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker SV,

Cruz-Correa M, Offerhaus JA. Very high risk of cancer in familial Peutz-Jeghers syndrome.

Gastroenterology 2000;119(6):1447-53.

221. Lim W, Olschwang S, Keller JJ, Westerman AM, Menko FH, Boardman LA, Scott RJ,

Trimbath J, Giardiello FM, Gruber SB, et al. Relative frequency and morphology of cancers in

STK11 mutation carriers. Gastroenterology 2004;126(7):1788-94.

222. Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S, Loukola A, Bignell G,

Warren W, Aminoff M, Hoglund P, et al. A serine/threonine kinase gene defective in Peutz-

Jeghers syndrome. Nature 1998;391(6663):184-7.

223. Aretz S, Stienen D, Uhlhaas S, Stolte M, Entius MM, Loff S, Back W, Kaufmann A,

Keller KM, Blaas SH, et al. High proportion of large genomic deletions and a genotype

phenotype update in 80 unrelated families with juvenile polyposis syndrome. J Med Genet

2007;44(11):702-9.

224. Zbuk KM, Eng C. Hamartomatous polyposis syndromes. Nat Clin Pract Gastroenterol

Hepatol 2007;4(9):492-502.

225. Lynch HT, de la Chapelle A. Hereditary colorectal cancer. N Engl J Med

2003;348(10):919-32.

226. Lynch HT, de la Chapelle A. Genetic susceptibility to non-polyposis colorectal cancer. J

Med Genet 1999;36(11):801-18.

227. Jass JR, Stewart SM. Evolution of hereditary non-polyposis colorectal cancer. Gut

1992;33(6):783-6.

228. Smyrk TC, Watson P, Kaul K, Lynch HT. Tumor-infiltrating lymphocytes are a marker

for microsatellite instability in colorectal carcinoma. Cancer 2001;91(12):2417-22.

Page 215: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

198

229. Jass JR, Do KA, Simms LA, Iino H, Wynter C, Pillay SP, Searle J, Radford-Smith G,

Young J, Leggett B. Morphology of sporadic colorectal cancer with DNA replication errors. Gut

1998;42(5):673-9.

230. Vasen HF, Mecklin JP, Khan PM, Lynch HT. The International Collaborative Group on

Hereditary Non-Polyposis Colorectal Cancer (ICG-HNPCC). Dis Colon Rectum

1991;34(5):424-5.

231. Vasen HF, Watson P, Mecklin JP, Lynch HT. New clinical criteria for hereditary

nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International

Collaborative group on HNPCC. Gastroenterology 1999;116(6):1453-6.

232. Rodriguez-Bigas MA, Boland CR, Hamilton SR, Henson DE, Jass JR, Khan PM, Lynch

H, Perucho M, Smyrk T, Sobin L, et al. A National Cancer Institute Workshop on Hereditary

Nonpolyposis Colorectal Cancer Syndrome: meeting highlights and Bethesda guidelines. J Natl

Cancer Inst 1997;89(23):1758-62.

233. Umar A, Risinger JI, Hawk ET, Barrett JC. Testing guidelines for hereditary non-

polyposis colorectal cancer. Nat Rev Cancer 2004;4(2):153-8.

234. Casey G, Lindor NM, Papadopoulos N, Thibodeau SN, Moskow J, Steelman S, Buzin

CH, Sommer SS, Collins CE, Butz M, et al. Conversion analysis for mutation detection in MLH1

and MSH2 in patients with colorectal cancer. Jama 2005;293(7):799-809.

235. Lynch J. The genetics and natural history of hereditary colon cancer. Semin Oncol Nurs

1997;13(2):91-8.

236. Nystrom-Lahti M, Wu Y, Moisio AL, Hofstra RM, Osinga J, Mecklin JP, Jarvinen HJ,

Leisti J, Buys CH, de la Chapelle A, et al. DNA mismatch repair gene mutations in 55 kindreds

with verified or putative hereditary non-polyposis colorectal cancer. Hum Mol Genet

1996;5(6):763-9.

237. Vogelstein BK, K.W., editor. The Genetic Basis of Human Cancer. New York: McGraw-

Hill; 1998.

238. Lindor NM, Rabe K, Petersen GM, Haile R, Casey G, Baron J, Gallinger S, Bapat B,

Aronson M, Hopper J, et al. Lower cancer incidence in Amsterdam-I criteria families without

mismatch repair deficiency: familial colorectal cancer type X. Jama 2005;293(16):1979-85.

239. Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J, Fishel R,

Lindor NM, Burgart LJ, Hamelin R, et al. Revised Bethesda Guidelines for hereditary

nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer

Inst 2004;96(4):261-8.

240. Watson P, Lynch HT. Extracolonic cancer in hereditary nonpolyposis colorectal cancer.

Cancer 1993;71(3):677-85.

Page 216: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

199

241. Aarnio M, Sankila R, Pukkala E, Salovaara R, Aaltonen LA, de la Chapelle A, Peltomaki

P, Mecklin JP, Jarvinen HJ. Cancer risk in mutation carriers of DNA-mismatch-repair genes. Int

J Cancer 1999;81(2):214-8.

242. Meyer LA, Broaddus RR, Lu KH. Endometrial cancer and Lynch syndrome: clinical and

pathologic considerations. Cancer Control 2009;16(1):14-22.

243. Hendriks YM, Wagner A, Morreau H, Menko F, Stormorken A, Quehenberger F,

Sandkuijl L, Moller P, Genuardi M, Van Houwelingen H, et al. Cancer risk in hereditary

nonpolyposis colorectal cancer due to MSH6 mutations: impact on counseling and surveillance.

Gastroenterology 2004;127(1):17-25.

244. Broaddus RR, Lynch HT, Chen LM, Daniels MS, Conrad P, Munsell MF, White KG,

Luthra R, Lu KH. Pathologic features of endometrial carcinoma associated with HNPCC: a

comparison with sporadic endometrial carcinoma. Cancer 2006;106(1):87-94.

245. Goodfellow PJ, Buttin BM, Herzog TJ, Rader JS, Gibb RK, Swisher E, Look K, Walls

KC, Fan MY, Mutch DG. Prevalence of defective DNA mismatch repair and MSH6 mutation in

an unselected series of endometrial cancers. Proc Natl Acad Sci U S A 2003;100(10):5908-13.

246. Esteller M, Levine R, Baylin SB, Ellenson LH, Herman JG. MLH1 promoter

hypermethylation is associated with the microsatellite instability phenotype in sporadic

endometrial carcinomas. Oncogene 1998;17(18):2413-7.

247. Mackay HJ, Gallinger S, Tsao MS, McLachlin CM, Tu D, Keiser K, Eisenhauer EA, Oza

AM. Prognostic value of microsatellite instability (MSI) and PTEN expression in women with

endometrial cancer: results from studies of the NCIC Clinical Trials Group (NCIC CTG). Eur J

Cancer 2010;46(8):1365-73.

248. Giardiello FM, Brensinger JD, Petersen GM, Luce MC, Hylind LM, Bacon JA, Booker

SV, Parker RD, Hamilton SR. The use and interpretation of commercial APC gene testing for

familial adenomatous polyposis. N Engl J Med 1997;336(12):823-7.

249. Eng C, Hampel H, de la Chapelle A. Genetic testing for cancer predisposition. Annu Rev

Med 2001;52:371-400.

250. Burke W, Petersen G, Lynch P, Botkin J, Daly M, Garber J, Kahn MJ, McTiernan A,

Offit K, Thomson E, et al. Recommendations for follow-up care of individuals with an inherited

predisposition to cancer. I. Hereditary nonpolyposis colon cancer. Cancer Genetics Studies

Consortium. JAMA 1997;277(11):915-9.

251. Jarvinen HJ, Aarnio M, Mustonen H, Aktan-Collan K, Aaltonen LA, Peltomaki P, De La

Chapelle A, Mecklin JP. Controlled 15-year trial on screening for colorectal cancer in families

with hereditary nonpolyposis colorectal cancer. Gastroenterology 2000;118(5):829-34.

252. Parker BO, Marinus MG. Repair of DNA heteroduplexes containing small heterologous

sequences in Escherichia coli. Proc Natl Acad Sci U S A 1992;89(5):1730-4.

Page 217: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

200

253. Harfe BD, Jinks-Robertson S. DNA mismatch repair and genetic instability. Annu Rev

Genet 2000;34:359-399.

254. Cooper DL, Lahue RS, Modrich P. Methyl-directed mismatch repair is bidirectional. J

Biol Chem 1993;268(16):11823-9.

255. Dao V, Modrich P. Mismatch-, MutS-, MutL-, and helicase II-dependent unwinding from

the single-strand break of an incised heteroduplex. J Biol Chem 1998;273(15):9202-7.

256. Ban C, Yang W. Crystal structure and ATPase activity of MutL: implications for DNA

repair and mutagenesis. Cell 1998;95(4):541-52.

257. Wu TH, Marinus MG. Deletion mutation analysis of the mutS gene in Escherichia coli. J

Biol Chem 1999;274(9):5948-52.

258. Hall MC, Matson SW. The Escherichia coli MutL protein physically interacts with MutH

and stimulates the MutH-associated endonuclease activity. J Biol Chem 1999;274(3):1306-12.

259. Hall MC, Jordan JR, Matson SW. Evidence for a physical interaction between the

Escherichia coli methyl-directed mismatch repair proteins MutL and UvrD. EMBO J

1998;17(5):1535-41.

260. Li GM, Modrich P. Restoration of mismatch repair to nuclear extracts of H6 colorectal

tumor cells by a heterodimer of human MutL homologs. Proc Natl Acad Sci U S A

1995;92(6):1950-4.

261. Raschle M, Marra G, Nystrom-Lahti M, Schar P, Jiricny J. Identification of hMutLbeta, a

heterodimer of hMLH1 and hPMS1. J Biol Chem 1999;274(45):32368-75.

262. Lipkin SM, Wang V, Jacoby R, Banerjee-Basu S, Baxevanis AD, Lynch HT, Elliott RM,

Collins FS. MLH3: a DNA mismatch repair gene associated with mammalian microsatellite

instability. Nat Genet 2000;24(1):27-35.

263. Lipkin SM, Moens PB, Wang V, Lenzi M, Shanmugarajah D, Gilgeous A, Thomas J,

Cheng J, Touchman JW, Green ED, et al. Meiotic arrest and aneuploidy in MLH3-deficient

mice. Nat Genet 2002;31(4):385-90.

264. Genschel J, Littman SJ, Drummond JT, Modrich P. Isolation of MutSbeta from human

cells and comparison of the mismatch repair specificities of MutSbeta and MutSalpha. J Biol

Chem 1998;273(31):19895-901.

265. Macpherson P, Humbert O, Karran P. Frameshift mismatch recognition by the human

MutS alpha complex. Mutat Res 1998;408(1):55-66.

266. Acharya S, Wilson T, Gradia S, Kane MF, Guerrette S, Marsischky GT, Kolodner R,

Fishel R. hMSH2 forms specific mispair-binding complexes with hMSH3 and hMSH6. Proc Natl

Acad Sci U S A 1996;93(24):13629-34.

Page 218: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

201

267. Marra G, Iaccarino I, Lettieri T, Roscilli G, Delmastro P, Jiricny J. Mismatch repair

deficiency associated with overexpression of the MSH3 gene. Proc Natl Acad Sci U S A

1998;95(15):8568-73.

268. Edelmann W, Cohen PE, Kneitz B, Winand N, Lia M, Heyer J, Kolodner R, Pollard JW,

Kucherlapati R. Mammalian MutS homologue 5 is required for chromosome pairing in meiosis.

Nat Genet 1999;21(1):123-7.

269. Paquis-Flucklinger V, Santucci-Darmanin S, Paul R, Saunieres A, Turc-Carel C,

Desnuelle C. Cloning and expression analysis of a meiosis-specific MutS homolog: the human

MSH4 gene. Genomics 1997;44(2):188-94.

270. Edelmann W, Yang K, Kuraguchi M, Heyer J, Lia M, Kneitz B, Fan K, Brown AM,

Lipkin M, Kucherlapati R. Tumorigenesis in Mlh1 and Mlh1/Apc1638N mutant mice. Cancer

Res 1999;59(6):1301-7.

271. Prolla TA, Baker SM, Harris AC, Tsao JL, Yao X, Bronner CE, Zheng B, Gordon M,

Reneker J, Arnheim N, et al. Tumour susceptibility and spontaneous mutation in mice deficient

in Mlh1, Pms1 and Pms2 DNA mismatch repair. Nat Genet 1998;18(3):276-9.

272. Chen PC, Dudley S, Hagen W, Dizon D, Paxton L, Reichow D, Yoon SR, Yang K,

Arnheim N, Liskay RM, et al. Contributions by MutL homologues Mlh3 and Pms2 to DNA

mismatch repair and tumor suppression in the mouse. Cancer Res 2005;65(19):8662-70.

273. Yao X, Buermeyer AB, Narayanan L, Tran D, Baker SM, Prolla TA, Glazer PM, Liskay

RM, Arnheim N. Different mutator phenotypes in Mlh1- versus Pms2-deficient mice. Proc Natl

Acad Sci U S A 1999;96(12):6850-5.

274. Jiricny J. The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol

2006;7(5):335-46.

275. Reitmair AH, Schmits R, Ewel A, Bapat B, Redston M, Mitri A, Waterhouse P,

Mittrucker HW, Wakeham A, Liu B, et al. MSH2 deficient mice are viable and susceptible to

lymphoid tumours. Nat Genet 1995;11(1):64-70.

276. Edelmann W, Yang K, Umar A, Heyer J, Lau K, Fan K, Liedtke W, Cohen PE, Kane

MF, Lipford JR, et al. Mutation in the mismatch repair gene Msh6 causes cancer susceptibility.

Cell 1997;91(4):467-77.

277. Edelmann W, Umar A, Yang K, Heyer J, Kucherlapati M, Lia M, Kneitz B, Avdievich E,

Fan K, Wong E, et al. The DNA mismatch repair genes Msh3 and Msh6 cooperate in intestinal

tumor suppression. Cancer Res 2000;60(4):803-7.

278. de Wind N, Dekker M, Claij N, Jansen L, van Klink Y, Radman M, Riggins G, van der

Valk M, van't Wout K, te Riele H. HNPCC-like cancer predisposition in mice through

simultaneous loss of Msh3 and Msh6 mismatch-repair protein functions. Nat Genet

1999;23(3):359-62.

Page 219: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

202

279. Modrich P, Lahue R. Mismatch repair in replication fidelity, genetic recombination, and

cancer biology. Annu Rev Biochem 1996;65:101-33.

280. Kadyrov FA, Dzantiev L, Constantin N, Modrich P. Endonucleolytic function of

MutLalpha in human mismatch repair. Cell 2006;126(2):297-308.

281. Wei K, Clark AB, Wong E, Kane MF, Mazur DJ, Parris T, Kolas NK, Russell R, Hou H,

Jr., Kneitz B, et al. Inactivation of Exonuclease 1 in mice results in DNA mismatch repair

defects, increased cancer susceptibility, and male and female sterility. Genes Dev

2003;17(5):603-14.

282. Dzantiev L, Constantin N, Genschel J, Iyer RR, Burgers PM, Modrich P. A defined

human system that supports bidirectional mismatch-provoked excision. Mol Cell 2004;15(1):31-

41.

283. Kleczkowska HE, Marra G, Lettieri T, Jiricny J. hMSH3 and hMSH6 interact with

PCNA and colocalize with it to replication foci. Genes Dev 2001;15(6):724-36.

284. Lau PJ, Kolodner RD. Transfer of the MSH2.MSH6 complex from proliferating cell

nuclear antigen to mispaired bases in DNA. J Biol Chem 2003;278(1):14-7.

285. Zhang Y, Yuan F, Presnell SR, Tian K, Gao Y, Tomkinson AE, Gu L, Li GM.

Reconstitution of 5'-directed human mismatch repair in a purified system. Cell 2005;122(5):693-

705.

286. Kunz C, Saito Y, Schar P. DNA Repair in mammalian cells: Mismatched repair:

variations on a theme. Cell Mol Life Sci 2009;66(6):1021-38.

287. Constantin N, Dzantiev L, Kadyrov FA, Modrich P. Human mismatch repair:

reconstitution of a nick-directed bidirectional reaction. J Biol Chem 2005;280(48):39752-61.

288. Lopez de Saro FJ, O'Donnell M. Interaction of the beta sliding clamp with MutS, ligase,

and DNA polymerase I. Proc Natl Acad Sci U S A 2001;98(15):8376-80.

289. Dufner P, Marra G, Raschle M, Jiricny J. Mismatch recognition and DNA-dependent

stimulation of the ATPase activity of hMutSalpha is abolished by a single mutation in the

hMSH6 subunit. J Biol Chem 2000;275(47):36550-5.

290. Gradia S, Subramanian D, Wilson T, Acharya S, Makhov A, Griffith J, Fishel R.

hMSH2-hMSH6 forms a hydrolysis-independent sliding clamp on mismatched DNA. Mol Cell

1999;3(2):255-61.

291. Blackwell LJ, Martik D, Bjornson KP, Bjornson ES, Modrich P. Nucleotide-promoted

release of hMutSalpha from heteroduplex DNA is consistent with an ATP-dependent

translocation mechanism. J Biol Chem 1998;273(48):32055-62.

292. Wang H, Hays JB. Mismatch repair in human nuclear extracts: effects of internal DNA-

hairpin structures between mismatches and excision-initiation nicks on mismatch correction and

mismatch-provoked excision. J Biol Chem 2003;278(31):28686-93.

Page 220: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

203

293. Peltomaki P. Deficient DNA mismatch repair: a common etiologic factor for colon

cancer. Hum Mol Genet 2001;10(7):735-40.

294. Jun SH, Kim TG, Ban C. DNA mismatch repair system. Classical and fresh roles. Febs J

2006;273(8):1609-19.

295. O'Brien V, Brown R. Signalling cell cycle arrest and cell death through the MMR

System. Carcinogenesis 2006;27(4):682-92.

296. Hawn MT, Umar A, Carethers JM, Marra G, Kunkel TA, Boland CR, Koi M. Evidence

for a connection between the mismatch repair system and the G2 cell cycle checkpoint. Cancer

Res 1995;55(17):3721-5.

297. Duckett DR, Bronstein SM, Taya Y, Modrich P. hMutSalpha- and hMutLalpha-

dependent phosphorylation of p53 in response to DNA methylator damage. Proc Natl Acad Sci

U S A 1999;96(22):12384-8.

298. Hickman MJ, Samson LD. Role of DNA mismatch repair and p53 in signaling induction

of apoptosis by alkylating agents. Proc Natl Acad Sci U S A 1999;96(19):10764-9.

299. Aquilina G, Crescenzi M, Bignami M. Mismatch repair, G(2)/M cell cycle arrest and

lethality after DNA damage. Carcinogenesis 1999;20(12):2317-26.

300. Moreland NJ, Illand M, Kim YT, Paul J, Brown R. Modulation of drug resistance

mediated by loss of mismatch repair by the DNA polymerase inhibitor aphidicolin. Cancer Res

1999;59(9):2102-6.

301. Abraham RT. The ATM-related kinase, hSMG-1, bridges genome and RNA surveillance

pathways. DNA Repair (Amst) 2004;3(8-9):919-25.

302. Bode AM, Dong Z. Post-translational modification of p53 in tumorigenesis. Nat Rev

Cancer 2004;4(10):793-805.

303. Adamson AW, Beardsley DI, Kim WJ, Gao Y, Baskaran R, Brown KD. Methylator-

induced, mismatch repair-dependent G2 arrest is activated through Chk1 and Chk2. Mol Biol

Cell 2005;16(3):1513-26.

304. Adamson AW, Kim WJ, Shangary S, Baskaran R, Brown KD. ATM is activated in

response to N-methyl-N'-nitro-N-nitrosoguanidine-induced DNA alkylation. J Biol Chem

2002;277(41):38222-9.

305. Caporali S, Falcinelli S, Starace G, Russo MT, Bonmassar E, Jiricny J, D'Atri S. DNA

damage induced by temozolomide signals to both ATM and ATR: role of the mismatch repair

system. Mol Pharmacol 2004;66(3):478-91.

306. Stojic L, Mojas N, Cejka P, Di Pietro M, Ferrari S, Marra G, Jiricny J. Mismatch repair-

dependent G2 checkpoint induced by low doses of SN1 type methylating agents requires the

ATR kinase. Genes Dev 2004;18(11):1331-44.

Page 221: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

204

307. Luo Y, Lin FT, Lin WC. ATM-mediated stabilization of hMutL DNA mismatch repair

proteins augments p53 activation during DNA damage. Mol Cell Biol 2004;24(14):6430-44.

308. Hirose Y, Katayama M, Stokoe D, Haas-Kogan DA, Berger MS, Pieper RO. The p38

mitogen-activated protein kinase pathway links the DNA mismatch repair system to the G2

checkpoint and to resistance to chemotherapeutic DNA-methylating agents. Mol Cell Biol

2003;23(22):8306-15.

309. de Wind N, Dekker M, van Rossum A, van der Valk M, te Riele H. Mouse models for

hereditary nonpolyposis colorectal cancer. Cancer Res 1998;58(2):248-55.

310. de Wind N, Dekker M, Berns A, Radman M, te Riele H. Inactivation of the mouse Msh2

gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and

predisposition to cancer. Cell 1995;82(2):321-30.

311. Cejka P, Stojic L, Mojas N, Russell AM, Heinimann K, Cannavo E, di Pietro M, Marra

G, Jiricny J. Methylation-induced G(2)/M arrest requires a full complement of the mismatch

repair protein hMLH1. Embo J 2003;22(9):2245-54.

312. Wedge SR, Porteus JK, May BL, Newlands ES. Potentiation of temozolomide and

BCNU cytotoxicity by O(6)-benzylguanine: a comparative study in vitro. Br J Cancer

1996;73(4):482-90.

313. Houghton PJ, Stewart CF, Cheshire PJ, Richmond LB, Kirstein MN, Poquette CA, Tan

M, Friedman HS, Brent TP. Antitumor activity of temozolomide combined with irinotecan is

partly independent of O6-methylguanine-DNA methyltransferase and mismatch repair

phenotypes in xenograft models. Clin Cancer Res 2000;6(10):4110-8.

314. D'Atri S, Tentori L, Lacal PM, Graziani G, Pagani E, Benincasa E, Zambruno G,

Bonmassar E, Jiricny J. Involvement of the mismatch repair system in temozolomide-induced

apoptosis. Mol Pharmacol 1998;54(2):334-41.

315. White JH, Lusnak K, Fogel S. Mismatch-specific post-meiotic segregation frequency in

yeast suggests a heteroduplex recombination intermediate. Nature 1985;315(6017):350-2.

316. Prolla TA, Christie DM, Liskay RM. Dual requirement in yeast DNA mismatch repair for

MLH1 and PMS1, two homologs of the bacterial mutL gene. Mol Cell Biol 1994;14(1):407-15.

317. Reenan RA, Kolodner RD. Characterization of insertion mutations in the Saccharomyces

cerevisiae MSH1 and MSH2 genes: evidence for separate mitochondrial and nuclear functions.

Genetics 1992;132(4):975-85.

318. Williamson MS, Game JC, Fogel S. Meiotic gene conversion mutants in Saccharomyces

cerevisiae. I. Isolation and characterization of pms1-1 and pms1-2. Genetics 1985;110(4):609-46.

319. Nicholson A, Hendrix M, Jinks-Robertson S, Crouse GF. Regulation of mitotic

homeologous recombination in yeast. Functions of mismatch repair and nucleotide excision

repair genes. Genetics 2000;154(1):133-46.

Page 222: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

205

320. Chen W, Jinks-Robertson S. The role of the mismatch repair machinery in regulating

mitotic and meiotic recombination between diverged sequences in yeast. Genetics

1999;151(4):1299-313.

321. Hunter N, Chambers SR, Louis EJ, Borts RH. The mismatch repair system contributes to

meiotic sterility in an interspecific yeast hybrid. Embo J 1996;15(7):1726-33.

322. Harfe BD, Jinks-Robertson S. Mismatch repair proteins and mitotic genome stability.

Mutat Res 2000;451(1-2):151-67.

323. Her C, Wu X, Wan W, Doggett NA. Identification and characterization of the mouse

MutS homolog 5: Msh5. Mamm Genome 1999;10(11):1054-61.

324. Edelmann W, Cohen PE, Kane M, Lau K, Morrow B, Bennett S, Umar A, Kunkel T,

Cattoretti G, Chaganti R, et al. Meiotic pachytene arrest in MLH1-deficient mice. Cell

1996;85(7):1125-34.

325. Baker SM, Plug AW, Prolla TA, Bronner CE, Harris AC, Yao X, Christie DM, Monell C,

Arnheim N, Bradley A, et al. Involvement of mouse Mlh1 in DNA mismatch repair and meiotic

crossing over. Nat Genet 1996;13(3):336-42.

326. Baker SM, Bronner CE, Zhang L, Plug AW, Robatzek M, Warren G, Elliott EA, Yu J,

Ashley T, Arnheim N, et al. Male mice defective in the DNA mismatch repair gene PMS2

exhibit abnormal chromosome synapsis in meiosis. Cell 1995;82(2):309-19.

327. Diaz M, Flajnik MF. Evolution of somatic hypermutation and gene conversion in

adaptive immunity. Immunol Rev 1998;162:13-24.

328. Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, Honjo T. Class switch

recombination and hypermutation require activation-induced cytidine deaminase (AID), a

potential RNA editing enzyme. Cell 2000;102(5):553-63.

329. Muramatsu M, Sankaranand VS, Anant S, Sugai M, Kinoshita K, Davidson NO, Honjo

T. Specific expression of activation-induced cytidine deaminase (AID), a novel member of the

RNA-editing deaminase family in germinal center B cells. J Biol Chem 1999;274(26):18470-6.

330. Peled JU, Kuang FL, Iglesias-Ussel MD, Roa S, Kalis SL, Goodman MF, Scharff MD.

The biochemistry of somatic hypermutation. Annu Rev Immunol 2008;26:481-511.

331. Rada C, Ehrenstein MR, Neuberger MS, Milstein C. Hot spot focusing of somatic

hypermutation in MSH2-deficient mice suggests two stages of mutational targeting. Immunity

1998;9(1):135-41.

332. Cascalho M, Wong J, Steinberg C, Wabl M. Mismatch repair co-opted by hypermutation.

Science 1998;279(5354):1207-10.

333. Winter DB, Phung QH, Umar A, Baker SM, Tarone RE, Tanaka K, Liskay RM, Kunkel

TA, Bohr VA, Gearhart PJ. Altered spectra of hypermutation in antibodies from mice deficient

for the DNA mismatch repair protein PMS2. Proc Natl Acad Sci U S A 1998;95(12):6953-8.

Page 223: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

206

334. Dutta R, Inouye M. GHKL, an emergent ATPase/kinase superfamily. Trends Biochem

Sci 2000;25(1):24-8.

335. Meyers M, Theodosiou M, Acharya S, Odegaard E, Wilson T, Lewis JE, Davis TW,

Wilson-Van Patten C, Fishel R, Boothman DA. Cell cycle regulation of the human DNA

mismatch repair genes hMSH2, hMLH1, and hPMS2. Cancer Res 1997;57(2):206-8.

336. Wu X, Platt JL, Cascalho M. Dimerization of MLH1 and PMS2 limits nuclear

localization of MutLalpha. Mol Cell Biol 2003;23(9):3320-8.

337. Brieger A, Plotz G, Raedle J, Weber N, Baum W, Caspary WF, Zeuzem S, Trojan J.

Characterization of the nuclear import of human MutLalpha. Mol Carcinog 2005;43(1):51-8.

338. Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, Qin J. BASC, a super complex of

BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures.

Genes Dev 2000;14(8):927-39.

339. Deng G, Chen A, Hong J, Chae HS, Kim YS. Methylation of CpG in a small region of

the hMLH1 promoter invariably correlates with the absence of gene expression. Cancer Res

1999;59(9):2029-33.

340. Capel E, Flejou JF, Hamelin R. Assessment of MLH1 promoter methylation in relation to

gene expression requires specific analysis. Oncogene 2007;26(54):7596-600.

341. Deng G, Chen A, Pong E, Kim YS. Methylation in hMLH1 promoter interferes with its

binding to transcription factor CBF and inhibits gene expression. Oncogene 2001;20(48):7120-7.

342. Lin JC, Jeong S, Liang G, Takai D, Fatemi M, Tsai YC, Egger G, Gal-Yam EN, Jones

PA. Role of nucleosomal occupancy in the epigenetic silencing of the MLH1 CpG island. Cancer

Cell 2007;12(5):432-44.

343. Ianzano L, Zhao XC, Minassian BA, Scherer SW. Identification of a novel protein

interacting with laforin, the EPM2a progressive myoclonus epilepsy gene product. Genomics

2003;81(6):579-87.

344. Papadopoulos N, Nicolaides NC, Wei YF, Ruben SM, Carter KC, Rosen CA, Haseltine

WA, Fleischmann RD, Fraser CM, Adams MD, et al. Mutation of a mutL homolog in hereditary

colon cancer. Science 1994;263(5153):1625-9.

345. Bronner CE, Baker SM, Morrison PT, Warren G, Smith LG, Lescoe MK, Kane M,

Earabino C, Lipford J, Lindblom A, et al. Mutation in the DNA mismatch repair gene

homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature

1994;368(6468):258-61.

346. Leach FS, Nicolaides NC, Papadopoulos N, Liu B, Jen J, Parsons R, Peltomaki P,

Sistonen P, Aaltonen LA, Nystrom-Lahti M, et al. Mutations of a mutS homolog in hereditary

nonpolyposis colorectal cancer. Cell 1993;75(6):1215-25.

Page 224: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

207

347. Fishel R, Lescoe MK, Rao MR, Copeland NG, Jenkins NA, Garber J, Kane M, Kolodner

R. The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis

colon cancer. Cell 1993;75(5):1027-38.

348. Lynch HT, Lynch PM, Pester J, Fusaro RM. The cancer family syndrome. Rare

cutaneous phenotypic linkage of Torre's syndrome. Arch Intern Med 1981;141(5):607-11.

349. Ponti G, Ponz de Leon M. Muir-Torre syndrome. Lancet Oncol 2005;6(12):980-7.

350. Bapat B, Xia L, Madlensky L, Mitri A, Tonin P, Narod SA, Gallinger S. The genetic

basis of Muir-Torre syndrome includes the hMLH1 locus. Am J Hum Genet 1996;59(3):736-9.

351. Kruse R, Lamberti C, Wang Y, Ruelfs C, Bruns A, Esche C, Lehmann P, Ruzicka T,

Rutten A, Friedl W, et al. Is the mismatch repair deficient type of Muir-Torre syndrome confined

to mutations in the hMSH2 gene? Hum Genet 1996;98(6):747-50.

352. Mangold E, Rahner N, Friedrichs N, Buettner R, Pagenstecher C, Aretz S, Friedl W,

Ruzicka T, Propping P, Rutten A, et al. MSH6 mutation in Muir-Torre syndrome: could this be a

rare finding? Br J Dermatol 2007;156(1):158-62.

353. Arnold A, Payne S, Fisher S, Fricker D, Soloway J, White SM, Novelli M, MacDonald

K, Mackay J, Groves R, et al. An individual with Muir-Torre syndrome found to have a

pathogenic MSH6 gene mutation. Fam Cancer 2007;6(3):317-21.

354. Perera S, Ramyar L, Mitri A, Pollett A, Gallinger S, Speevak MD, Aronson M, Bapat B.

A novel complex mutation in MSH2 contributes to both Muir-Torre and Lynch Syndrome. J

Hum Genet 2009;55(1):37-41.

355. De Vos M, Hayward BE, Picton S, Sheridan E, Bonthron DT. Novel PMS2 pseudogenes

can conceal recessive mutations causing a distinctive childhood cancer syndrome. Am J Hum

Genet 2004;74(5):954-64.

356. Gallinger S, Aronson M, Shayan K, Ratcliffe EM, Gerstle JT, Parkin PC, Rothenmund H,

Croitoru M, Baumann E, Durie PR, et al. Gastrointestinal cancers and neurofibromatosis type 1

features in children with a germline homozygous MLH1 mutation. Gastroenterology

2004;126(2):576-85.

357. Menko FH, Kaspers GL, Meijer GA, Claes K, van Hagen JM, Gille JJ. A homozygous

MSH6 mutation in a child with cafe-au-lait spots, oligodendroglioma and rectal cancer. Fam

Cancer 2004;3(2):123-7.

358. Trimbath JD, Petersen GM, Erdman SH, Ferre M, Luce MC, Giardiello FM. Cafe-au-lait

spots and early onset colorectal neoplasia: a variant of HNPCC? Fam Cancer 2001;1(2):101-5.

359. Whiteside D, McLeod R, Graham G, Steckley JL, Booth K, Somerville MJ, Andrew SE.

A homozygous germ-line mutation in the human MSH2 gene predisposes to hematological

malignancy and multiple cafe-au-lait spots. Cancer Res 2002;62(2):359-62.

Page 225: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

208

360. De Vos M, Hayward BE, Charlton R, Taylor GR, Glaser AW, Picton S, Cole TR, Maher

ER, McKeown CM, Mann JR, et al. PMS2 mutations in childhood cancer. J Natl Cancer Inst

2006;98(5):358-61.

361. Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell SM, Krush AJ, Berk T,

Cohen Z, Tetu B, et al. The molecular basis of Turcot's syndrome. N Engl J Med

1995;332(13):839-47.

362. De Rosa M, Fasano C, Panariello L, Scarano MI, Belli G, Iannelli A, Ciciliano F, Izzo P.

Evidence for a recessive inheritance of Turcot's syndrome caused by compound heterozygous

mutations within the PMS2 gene. Oncogene 2000;19(13):1719-23.

363. Monzon JG, Cremin C, Armstrong L, Nuk J, Young S, Horsman DE, Garbutt K, Bajdik

CD, Gill S. Validation of predictive models for germline mutations in DNA mismatch repair

genes in colorectal cancer. Int J Cancer 2010;126(4):930-9.

364. Kang HH, Williams R, Leary J, Ringland C, Kirk J, Ward R. Evaluation of models to

predict BRCA germline mutations. Br J Cancer 2006;95(7):914-20.

365. Niendorf KB, Goggins W, Yang G, Tsai KY, Shennan M, Bell DW, Sober AJ, Hogg D,

Tsao H. MELPREDICT: a logistic regression model to estimate CDKN2A carrier probability. J

Med Genet 2006;43(6):501-6.

366. Wang W, Chen S, Brune KA, Hruban RH, Parmigiani G, Klein AP. PancPRO: risk

assessment for individuals with a family history of pancreatic cancer. J Clin Oncol

2007;25(11):1417-22.

367. Wijnen JT, Vasen HF, Khan PM, Zwinderman AH, van der Klift H, Mulder A, Tops C,

Moller P, Fodde R. Clinical findings with implications for genetic testing in families with

clustering of colorectal cancer. N Engl J Med 1998;339(8):511-8.

368. Chen S, Wang W, Lee S, Nafa K, Lee J, Romans K, Watson P, Gruber SB, Euhus D,

Kinzler KW, et al. Prediction of germline mutations and cancer risk in the Lynch syndrome.

Jama 2006;296(12):1479-87.

369. Balmana J, Stockwell DH, Steyerberg EW, Stoffel EM, Deffenbaugh AM, Reid JE, Ward

B, Scholl T, Hendrickson B, Tazelaar J, et al. Prediction of MLH1 and MSH2 mutations in

Lynch syndrome. Jama 2006;296(12):1469-78.

370. Barnetson RA, Tenesa A, Farrington SM, Nicholl ID, Cetnarskyj R, Porteous ME,

Campbell H, Dunlop MG. Identification and survival of carriers of mutations in DNA mismatch-

repair genes in colon cancer. N Engl J Med 2006;354(26):2751-63.

371. Pouchet CJ, Wong N, Chong G, Sheehan MJ, Schneider G, Rosen-Sheidley B, Foulkes

W, Tischkowitz M. A comparison of models used to predict MLH1, MSH2 and MSH6 mutation

carriers. Ann Oncol 2009;20(4):681-8.

Page 226: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

209

372. Woods MO, Williams P, Careen A, Edwards L, Bartlett S, McLaughlin JR,

Younghusband HB. A new variant database for mismatch repair genes associated with Lynch

syndrome. Hum Mutat 2007;28(7):669-673.

373. Suter CM, Martin DI, Ward RL. Germline epimutation of MLH1 in individuals with

multiple cancers. Nat Genet 2004;36(5):497-501.

374. Earle JS, Luthra R, Romans A, Abraham R, Ensor J, Yao H, Hamilton SR. Association of

MicroRNA Expression with Microsatellite Instability Status in Colorectal Adenocarcinoma. J

Mol Diagn 2010.

375. Ligtenberg MJ, Kuiper RP, Chan TL, Goossens M, Hebeda KM, Voorendt M, Lee TY,

Bodmer D, Hoenselaar E, Hendriks-Cornelissen SJ, et al. Heritable somatic methylation and

inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3' exons of

TACSTD1. Nat Genet 2009;41(1):112-7.

376. Niessen RC, Hofstra RM, Westers H, Ligtenberg MJ, Kooi K, Jager PO, de Groote ML,

Dijkhuizen T, Olderode-Berends MJ, Hollema H, et al. Germline hypermethylation of MLH1

and EPCAM deletions are a frequent cause of Lynch syndrome. Genes Chromosomes Cancer

2009;48(8):737-44.

377. Nagasaka T, Rhees J, Kloor M, Gebert J, Naomoto Y, Boland CR, Goel A. Somatic

hypermethylation of MSH2 is a frequent event in Lynch Syndrome colorectal cancers. Cancer

Res 2010;70(8):3098-108.

378. Jacob S, Praz F. DNA mismatch repair defects: role in colorectal carcinogenesis.

Biochimie 2002;84(1):27-47.

379. Drotschmann K, Topping RP, Clodfelter JE, Salsbury FR. Mutations in the nucleotide-

binding domain of MutS homologs uncouple cell death from cell survival. DNA Repair (Amst)

2004;3(7):729-42.

380. Kariola R, Hampel H, Frankel WL, Raevaara TE, de la Chapelle A, Nystrom-Lahti M.

MSH6 missense mutations are often associated with no or low cancer susceptibility. Br J Cancer

2004;91(7):1287-92.

381. Lin DP, Wang Y, Scherer SJ, Clark AB, Yang K, Avdievich E, Jin B, Werling U, Parris

T, Kurihara N, et al. An Msh2 point mutation uncouples DNA mismatch repair and apoptosis.

Cancer Res 2004;64(2):517-22.

382. Lipkin SM, Rozek LS, Rennert G, Yang W, Chen PC, Hacia J, Hunt N, Shin B, Fodor S,

Kokoris M, et al. The MLH1 D132H variant is associated with susceptibility to sporadic

colorectal cancer. Nat Genet 2004;36(7):694-9.

383. Raevaara TE, Korhonen MK, Lohi H, Hampel H, Lynch E, Lonnqvist KE, Holinski-

Feder E, Sutter C, McKinnon W, Duraisamy S, et al. Functional significance and clinical

phenotype of nontruncating mismatch repair variants of MLH1. Gastroenterology

2005;129(2):537-49.

Page 227: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

210

384. Shimodaira H, Filosi N, Shibata H, Suzuki T, Radice P, Kanamaru R, Friend SH,

Kolodner RD, Ishioka C. Functional analysis of human MLH1 mutations in Saccharomyces

cerevisiae. Nat Genet 1998;19(4):384-9.

385. Trojan J, Zeuzem S, Randolph A, Hemmerle C, Brieger A, Raedle J, Plotz G, Jiricny J,

Marra G. Functional analysis of hMLH1 variants and HNPCC-related mutations using a human

expression system. Gastroenterology 2002;122(1):211-9.

386. Chan PA, Duraisamy S, Miller PJ, Newell JA, McBride C, Bond JP, Raevaara T, Ollila

S, Nystrom M, Grimm AJ, et al. Interpreting missense variants: comparing computational

methods in human disease genes CDKN2A, MLH1, MSH2, MECP2, and tyrosinase (TYR).

Hum Mutat 2007;28(7):683-693.

387. Pang Q, Prolla TA, Liskay RM. Functional domains of the Saccharomyces cerevisiae

Mlh1p and Pms1p DNA mismatch repair proteins and their relevance to human hereditary

nonpolyposis colorectal cancer-associated mutations. Mol Cell Biol 1997;17(8):4465-73.

388. Guerrette S, Acharya S, Fishel R. The interaction of the human MutL homologues in

hereditary nonpolyposis colon cancer. J Biol Chem 1999;274(10):6336-41.

389. Perera S, Bapat B. The MLH1 variants p.Arg265Cys and p.Lys618Ala affect protein

stability while p.Leu749Gln affects heterodimer formation. Hum Mutat 2008;29(2):332.

390. Brieger A, Trojan J, Raedle J, Plotz G, Zeuzem S. Transient mismatch repair gene

transfection for functional analysis of genetic hMLH1 and hMSH2 variants. Gut 2002;51(5):677-

84.

391. Raevaara TE, Gerdes AM, Lonnqvist KE, Tybjaerg-Hansen A, Abdel-Rahman WM,

Kariola R, Peltomaki P, Nystrom-Lahti M. HNPCC mutation MLH1 P648S makes the functional

protein unstable, and homozygosity predisposes to mild neurofibromatosis type 1. Genes

Chromosomes Cancer 2004;40(3):261-5.

392. Nystrom-Lahti M, Perrera C, Raschle M, Panyushkina-Seiler E, Marra G, Curci A,

Quaresima B, Costanzo F, D'Urso M, Venuta S, et al. Functional analysis of MLH1 mutations

linked to hereditary nonpolyposis colon cancer. Genes Chromosomes Cancer 2002;33(2):160-7.

393. Modrich P. Mechanisms in eukaryotic mismatch repair. J Biol Chem

2006;281(41):30305-9.

394. Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA. DNA repair pathways as

targets for cancer therapy. Nat Rev Cancer 2008;8(3):193-204.

395. Li C, Bapat B, Alman BA. Adenomatous polyposis coli gene mutation alters proliferation

through its beta-catenin-regulatory function in aggressive fibromatosis (desmoid tumor). Am J

Pathol 1998;153(3):709-14.

396. Yuan ZQ, Gottlieb B, Beitel LK, Wong N, Gordon PH, Wang Q, Puisieux A, Foulkes

WD, Trifiro M. Polymorphisms and HNPCC: PMS2-MLH1 protein interactions diminished by

single nucleotide polymorphisms. Hum Mutat 2002;19(2):108-13.

Page 228: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

211

397. Chung DC, Rustgi AK. The hereditary nonpolyposis colorectal cancer syndrome:

genetics and clinical implications. Ann Intern Med 2003;138(7):560-70.

398. Andrew SE, Whiteside D, Buzin C, Greenberg C, Spriggs E. An intronic polymorphism

of the hMLH1 gene contributes toward incomplete genetic testing for HNPCC. Genet Test

2002;6(4):319-22.

399. Kurzawski G, Suchy J, Kladny J, Safranow K, Jakubowska A, Elsakov P, Kucinskas V,

Gardovski J, Irmejs A, Sibul H, et al. Germline MSH2 and MLH1 mutational spectrum in

HNPCC families from Poland and the Baltic States. J Med Genet 2002;39(10):E65.

400. Blasi MF, Ventura I, Aquilina G, Degan P, Bertario L, Bassi C, Radice P, Bignami M. A

human cell-based assay to evaluate the effects of alterations in the MLH1 mismatch repair gene.

Cancer Res 2006;66(18):9036-44.

401. Ellison AR, Lofing J, Bitter GA. Functional analysis of human MLH1 and MSH2

missense variants and hybrid human-yeast MLH1 proteins in Saccharomyces cerevisiae. Hum

Mol Genet 2001;10(18):1889-900.

402. Fearnhead NS, Wilding JL, Winney B, Tonks S, Bartlett S, Bicknell DC, Tomlinson IP,

Mortensen NJ, Bodmer WF. Multiple rare variants in different genes account for multifactorial

inherited susceptibility to colorectal adenomas. Proc Natl Acad Sci U S A 2004;101(45):15992-

7.

403. Kondo E, Suzuki H, Horii A, Fukushige S. A yeast two-hybrid assay provides a simple

way to evaluate the vast majority of hMLH1 germ-line mutations. Cancer Res

2003;63(12):3302-8.

404. Mohd AB, Palama B, Nelson SE, Tomer G, Nguyen M, Huo X, Buermeyer AB.

Truncation of the C-terminus of human MLH1 blocks intracellular stabilization of PMS2 and

disrupts DNA mismatch repair. DNA Repair (Amst) 2006;5(3):347-61.

405. Koi M, Umar A, Chauhan DP, Cherian SP, Carethers JM, Kunkel TA, Boland CR.

Human chromosome 3 corrects mismatch repair deficiency and microsatellite instability and

reduces N-methyl-N'-nitro-N-nitrosoguanidine tolerance in colon tumor cells with homozygous

hMLH1 mutation. Cancer Res 1994;54(16):4308-12.

406. Ramensky V, Bork P, Sunyaev S. Human non-synonymous SNPs: server and survey.

Nucleic Acids Res 2002;30(17):3894-900.

407. Ng PC, Henikoff S. Predicting deleterious amino acid substitutions. Genome Res

2001;11(5):863-74.

408. Charames GS, Ramyar L, Mitri A, Berk T, Cheng H, Jung J, Bocangel P, Chodirker B,

Greenberg C, Spriggs E, et al. A large novel deletion in the APC promoter region causes gene

silencing and leads to classical familial adenomatous polyposis in a Manitoba Mennonite

kindred. Hum Genet 2008;124(5):535-41.

Page 229: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

212

409. Esufali S, Bapat B. Cross-talk between Rac1 GTPase and dysregulated Wnt signaling

pathway leads to cellular redistribution of beta-catenin and TCF/LEF-mediated transcriptional

activation. Oncogene 2004;23(50):8260-71.

410. Woods MO, Hyde AJ, Curtis FK, Stuckless S, Green JS, Pollett AF, Robb JD, Green RC,

Croitoru ME, Careen A, et al. High frequency of hereditary colorectal cancer in Newfoundland

likely involves novel susceptibility genes. Clin Cancer Res 2005;11(19 Pt 1):6853-61.

411. Colombino M, Cossu A, Arba A, Manca A, Curci A, Avallone A, Comella G, Botti G,

Scintu F, Amoruso M, et al. Microsatellite instability and mutation analysis among southern

Italian patients with colorectal carcinoma: detection of different alterations accounting for MLH1

and MSH2 inactivation in familial cases. Ann Oncol 2003;14(10):1530-6.

412. Losi L, Di Gregorio C, Pedroni M, Ponti G, Roncucci L, Scarselli A, Genuardi M,

Baglioni S, Marino M, Rossi G, et al. Molecular genetic alterations and clinical features in early-

onset colorectal carcinomas and their role for the recognition of hereditary cancer syndromes.

Am J Gastroenterol 2005;100(10):2280-7.

413. Takahashi M, Shimodaira H, Andreutti-Zaugg C, Iggo R, Kolodner RD, Ishioka C.

Functional analysis of human MLH1 variants using yeast and in vitro mismatch repair assays.

Cancer Res 2007;67(10):4595-604.

414. Boyer JC, Umar A, Risinger JI, Lipford JR, Kane M, Yin S, Barrett JC, Kolodner RD,

Kunkel TA. Microsatellite instability, mismatch repair deficiency, and genetic defects in human

cancer cell lines. Cancer Res 1995;55(24):6063-70.

415. Genuardi M, Viel A, Bonora D, Capozzi E, Bellacosa A, Leonardi F, Valle R, Ventura A,

Pedroni M, Boiocchi M, et al. Characterization of MLH1 and MSH2 alternative splicing and its

relevance to molecular testing of colorectal cancer susceptibility. Hum Genet 1998;102(1):15-20.

416. Liu L, Markowitz S, Gerson SL. Mismatch repair mutations override alkyltransferase in

conferring resistance to temozolomide but not to 1,3-bis(2-chloroethyl)nitrosourea. Cancer Res

1996;56(23):5375-9.

417. Shcherbakova PV, Kunkel TA. Mutator phenotypes conferred by MLH1 overexpression

and by heterozygosity for mlh1 mutations. Mol Cell Biol 1999;19(4):3177-83.

418. Barnetson RA, Cartwright N, van Vliet A, Haq N, Drew K, Farrington S, Williams N,

Warner J, Campbell H, Porteous ME, et al. Classification of ambiguous mutations in DNA

mismatch repair genes identified in a population-based study of colorectal cancer. Hum Mutat

2008;29(3):367-74.

419. Pinsonneault J, Nielsen CU, Sadee W. Genetic variants of the human H+/dipeptide

transporter PEPT2: analysis of haplotype functions. J Pharmacol Exp Ther 2004;311(3):1088-96.

420. Zhang Y, Wang D, Johnson AD, Papp AC, Sadee W. Allelic expression imbalance of

human mu opioid receptor (OPRM1) caused by variant A118G. J Biol Chem

2005;280(38):32618-24.

Page 230: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

213

421. Murakami Y, Isogai K, Tomita H, Sakurai-Yageta M, Maruyama T, Hidaka A, Nose K,

Sugano K, Kaneko A. Detection of allelic imbalance in the gene expression of hMSH2 or RB1 in

lymphocytes from pedigrees of hereditary, nonpolyposis, colorectal cancer and retinoblastoma

by an RNA difference plot. J Hum Genet 2004;49(11):635-41.

422. Soravia C, Sugg SL, Berk T, Mitri A, Cheng H, Gallinger S, Cohen Z, Asa SL, Bapat

BV. Familial adenomatous polyposis-associated thyroid cancer: a clinical, pathological, and

molecular genetics study. Am J Pathol 1999;154(1):127-35.

423. Ge B, Gurd S, Gaudin T, Dore C, Lepage P, Harmsen E, Hudson TJ, Pastinen T. Survey

of allelic expression using EST mining. Genome Res 2005;15(11):1584-91.

424. Lee PD, Ge B, Greenwood CM, Sinnett D, Fortin Y, Brunet S, Fortin A, Takane M,

Skamene E, Pastinen T, et al. Mapping cis-acting regulatory variation in recombinant congenic

strains. Physiol Genomics 2006;25(2):294-302.

425. Cartegni L, Wang J, Zhu Z, Zhang MQ, Krainer AR. ESEfinder: A web resource to

identify exonic splicing enhancers. Nucleic Acids Res 2003;31(13):3568-71.

426. Smith PJ, Zhang C, Wang J, Chew SL, Zhang MQ, Krainer AR. An increased specificity

score matrix for the prediction of SF2/ASF-specific exonic splicing enhancers. Hum Mol Genet

2006;15(16):2490-508.

427. Renkonen E, Zhang Y, Lohi H, Salovaara R, Abdel-Rahman WM, Nilbert M, Aittomaki

K, Jarvinen HJ, Mecklin JP, Lindblom A, et al. Altered expression of MLH1, MSH2, and MSH6

in predisposition to hereditary nonpolyposis colorectal cancer. J Clin Oncol 2003;21(19):3629-

37.

428. Renkonen ET, Nieminen P, Abdel-Rahman WM, Moisio AL, Jarvela I, Arte S, Jarvinen

HJ, Peltomaki P. Adenomatous polyposis families that screen APC mutation-negative by

conventional methods are genetically heterogeneous. J Clin Oncol 2005;23(24):5651-9.

429. Lastella P, Surdo NC, Resta N, Guanti G, Stella A. In silico and in vivo splicing analysis

of MLH1 and MSH2 missense mutations shows exon- and tissue-specific effects. BMC

Genomics 2006;7:243.

430. Tournier I, Vezain M, Martins A, Charbonnier F, Baert-Desurmont S, Olschwang S,

Wang Q, Buisine MP, Soret J, Tazi J, et al. A large fraction of unclassified variants of the

mismatch repair genes MLH1 and MSH2 is associated with splicing defects. Hum Mutat

2008;29(12):1412-24.

431. Mecklin JP, Aarnio M, Laara E, Kairaluoma MV, Pylvanainen K, Peltomaki P, Aaltonen

LA, Jarvinen HJ. Development of colorectal tumors in colonoscopic surveillance in Lynch

syndrome. Gastroenterology 2007;133(4):1093-8.

432. Tournier I, Raux G, Di Fiore F, Marechal I, Leclerc C, Martin C, Wang Q, Buisine MP,

Stoppa-Lyonnet D, Olschwang S, et al. Analysis of the allele-specific expression of the

mismatch repair gene MLH1 using a simple DHPLC-Based Method. Hum Mutat

2004;23(4):379-84.

Page 231: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

214

433. Plotz G, Welsch C, Giron-Monzon L, Friedhoff P, Albrecht M, Piiper A, Biondi RM,

Lengauer T, Zeuzem S, Raedle J. Mutations in the MutSalpha interaction interface of MLH1 can

abolish DNA mismatch repair. Nucleic Acids Res 2006;34(22):6574-86.

434. Drost M, Zonneveld JB, van Dijk L, Morreau H, Tops CM, Vasen HF, Wijnen JT, de

Wind N. A cell-free assay for the functional analysis of variants of the mismatch repair protein

MLH1. Hum Mutat 2010;31(3):247-53.

435. de Leon MP, Benatti P, Di Gregorio C, Losi L, Pedroni M, Ponti G, Genuardi M, Viel A,

Lucci-Cordisco E, Rossi G, et al. Genotype-phenotype correlations in individuals with a founder

mutation in the MLH1 gene and hereditary non-polyposis colorectal cancer. Scand J

Gastroenterol 2007;42(6):746-53.

436. Peltomaki P, Vasen HF. Mutations predisposing to hereditary nonpolyposis colorectal

cancer: database and results of a collaborative study. The International Collaborative Group on

Hereditary Nonpolyposis Colorectal Cancer. Gastroenterology 1997;113(4):1146-58.

437. Frischmeyer PA, Dietz HC. Nonsense-mediated mRNA decay in health and disease. Hum

Mol Genet 1999;8(10):1893-900.

438. Sharp A, Pichert G, Lucassen A, Eccles D. RNA analysis reveals splicing mutations and

loss of expression defects in MLH1 and BRCA1. Hum Mutat 2004;24(3):272.

439. Gorlov IP, Gorlova OY, Frazier ML, Amos CI. Missense mutations in hMLH1 and

hMSH2 are associated with exonic splicing enhancers. Am J Hum Genet 2003;73(5):1157-61.

440. Distelman-Menachem T, Shapira T, Laitman Y, Kaufman B, Barak F, Tavtigian S,

Friedman E. Analysis of BRCA1/BRCA2 genes' contribution to breast cancer susceptibility in

high risk Jewish Ashkenazi women. Fam Cancer 2008.

441. Schofield MJ, Hsieh P. DNA mismatch repair: molecular mechanisms and biological

function. Annu Rev Microbiol 2003;57:579-608.

442. Lu HK, Broaddus RR. Gynecologic Cancers in Lynch Syndrome/HNPCC. Fam Cancer

2005;4(3):249-54.

443. Umar A. Lynch syndrome (HNPCC) and microsatellite instability. Dis Markers

2004;20(4-5):179-80.

444. Poynter JN, Siegmund KD, Weisenberger DJ, Long TI, Thibodeau SN, Lindor N, Young

J, Jenkins MA, Hopper JL, Baron JA, et al. Molecular characterization of MSI-H colorectal

cancer by MLHI promoter methylation, immunohistochemistry, and mismatch repair germline

mutation screening. Cancer Epidemiol Biomarkers Prev 2008;17(11):3208-15.

445. Hitchins MP, Lin VA, Buckle A, Cheong K, Halani N, Ku S, Kwok CT, Packham D,

Suter CM, Meagher A, et al. Epigenetic inactivation of a cluster of genes flanking MLH1 in

microsatellite-unstable colorectal cancer. Cancer Res 2007;67(19):9107-16.

Page 232: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

215

446. Raptis S, Mrkonjic M, Green RC, Pethe VV, Monga N, Chan YM, Daftary D, Dicks E,

Younghusband BH, Parfrey PS, et al. MLH1 -93G>A promoter polymorphism and the risk of

microsatellite-unstable colorectal cancer. J Natl Cancer Inst 2007;99(6):463-74.

447. Yu JH, Bigler J, Whitton J, Potter JD, Ulrich CM. Mismatch repair polymorphisms and

colorectal polyps: hMLH1-93G>A variant modifies risk associated with smoking. Am J

Gastroenterol 2006;101(6):1313-9.

448. Campbell PT, Curtin K, Ulrich C, Samowitz W, Bigler J, Velicer C, Caan B, Potter J,

Slattery M. Mismatch repair polymorphisms and risk of colon cancer, tumor microsatellite

instability, and interactions with lifestyle factors. Gut 2008.

449. Allan JM, Shorto J, Adlard J, Bury J, Coggins R, George R, Katory M, Quirke P,

Richman S, Scott D, et al. MLH1 -93G>A promoter polymorphism and risk of mismatch repair

deficient colorectal cancer. Int J Cancer 2008;123(10):2456-9.

450. Beiner ME, Rosen B, Fyles A, Harley I, Pal T, Siminovitch K, Zhang S, Sun P, Narod

SA. Endometrial cancer risk is associated with variants of the mismatch repair genes MLH1 and

MSH2. Cancer Epidemiol Biomarkers Prev 2006;15(9):1636-40.

451. Harley I, Rosen B, Risch HA, Siminovitch K, Beiner ME, McLaughlin J, Sun P, Narod

SA. Ovarian cancer risk is associated with a common variant in the promoter sequence of the

mismatch repair gene MLH1. Gynecol Oncol 2008;109(3):384-7.

452. Park SH, Lee GY, Jeon HS, Lee SJ, Kim KM, Jang SS, Kim CH, Lee WK, Kam S, Park

RW, et al. -93G-->A polymorphism of hMLH1 and risk of primary lung cancer. Int J Cancer

2004;112(4):678-82.

453. Ito E, Yanagisawa Y, Iwahashi Y, Suzuki Y, Nagasaki H, Akiyama Y, Sugano S, Yuasa

Y, Maruyama K. A core promoter and a frequent single-nucleotide polymorphism of the

mismatch repair gene hMLH1. Biochem Biophys Res Commun 1999;256(3):488-94.

454. Zhong X, Arita M, Koike J, Tsujita K, Hemmi H. A Single Nucleotide Polymorphism in

the Promoter Region of the hMLH1 Gene: Effect on Transcriptional Activity and Application as

a Marker for Detecting Allelic Loss. Journal of the Medical Society of Toho University

2001;48(2):114-124.

455. Evan G. Regulation of gene expression. Br Med Bull 1991;47(1):116-35.

456. Arita M, Zhong X, Min Z, Hemmi H, Shimatake H. Multiple sites required for expression

in 5'-flanking region of the hMLH1 gene. Gene 2003;306:57-65.

457. Walker LC, Waddell N, Ten Haaf A, Grimmond S, Spurdle AB. Use of expression data

and the CGEMS genome-wide breast cancer association study to identify genes that may modify

risk in BRCA1/2 mutation carriers. Breast Cancer Res Treat 2008;112(2):229-36.

458. Wang Y, Liu Y, Wu C, Zhang H, Zheng X, Zheng Z, Geiger TL, Nuovo GJ, Liu Y,

Zheng P. Epm2a suppresses tumor growth in an immunocompromised host by inhibiting Wnt

signaling. Cancer Cell 2006;10(3):179-90.

Page 233: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

216

459. Kako K, Wakamatsu H, Hamada T, Banasik M, Ohata K, Niki-Kuroiwa T, Suzuki S,

Takeuchi J, Ishida N. Examination of DNA-binding activity of neuronal transcription factors by

electrophoretical mobility shift assay. Brain Res Brain Res Protoc 1998;2(4):243-9.

460. Mac Partlin M, Homer E, Robinson H, McCormick CJ, Crouch DH, Durant ST,

Matheson EC, Hall AG, Gillespie DA, Brown R. Interactions of the DNA mismatch repair

proteins MLH1 and MSH2 with c-MYC and MAX. Oncogene 2003;22(6):819-25.

461. Bindra RS, Glazer PM. Co-repression of mismatch repair gene expression by hypoxia in

cancer cells: role of the Myc/Max network. Cancer Lett 2007;252(1):93-103.

462. Nakamura H, Tanimoto K, Hiyama K, Yunokawa M, Kawamoto T, Kato Y, Yoshiga K,

Poellinger L, Hiyama E, Nishiyama M. Human mismatch repair gene, MLH1, is transcriptionally

repressed by the hypoxia-inducible transcription factors, DEC1 and DEC2. Oncogene

2008;27(30):4200-9.

463. Bindra RS, Glazer PM. Genetic instability and the tumor microenvironment: towards the

concept of microenvironment-induced mutagenesis. Mutat Res 2005;569(1-2):75-85.

464. Chen H, Taylor NP, Sotamaa KM, Mutch DG, Powell MA, Schmidt AP, Feng S, Hampel

HL, de la Chapelle A, Goodfellow PJ. Evidence for heritable predisposition to epigenetic

silencing of MLH1. Int J Cancer 2007;120(8):1684-8.

465. Samowitz WS, Curtin K, Wolff RK, Albertsen H, Sweeney C, Caan BJ, Ulrich CM,

Potter JD, Slattery ML. The MLH1 -93 G>A promoter polymorphism and genetic and epigenetic

alterations in colon cancer. Genes Chromosomes Cancer 2008;47(10):835-44.

466. Deng G, Peng E, Gum J, Terdiman J, Sleisenger M, Kim YS. Methylation of hMLH1

promoter correlates with the gene silencing with a region-specific manner in colorectal cancer.

Br J Cancer 2002;86(4):574-9.

467. Chang DK, Ricciardiello L, Goel A, Chang CL, Boland CR. Steady-state regulation of

the human DNA mismatch repair system. J Biol Chem 2000;275(24):18424-31.

468. Cannavo E, Marra G, Sabates-Bellver J, Menigatti M, Lipkin SM, Fischer F, Cejka P,

Jiricny J. Expression of the MutL homologue hMLH3 in human cells and its role in DNA

mismatch repair. Cancer Res 2005;65(23):10759-66.

469. Korhonen MK, Raevaara TE, Lohi H, Nystrom M. Conditional nuclear localization of

hMLH3 suggests a minor activity in mismatch repair and supports its role as a low-risk gene in

HNPCC. Oncol Rep 2007;17(2):351-4.

470. Poser I, Sarov M, Hutchins JR, Heriche JK, Toyoda Y, Pozniakovsky A, Weigl D,

Nitzsche A, Hegemann B, Bird AW, et al. BAC TransgeneOmics: a high-throughput method for

exploration of protein function in mammals. Nat Methods 2008;5(5):409-15.

471. Campbell PT, Curtin K, Ulrich CM, Samowitz WS, Bigler J, Velicer CM, Caan B, Potter

JD, Slattery ML. Mismatch repair polymorphisms and risk of colon cancer, tumour microsatellite

instability and interactions with lifestyle factors. Gut 2009;58(5):661-7.

Page 234: Assessment of missense alterations in MLH1 and their ......Pethe, Miralem Mrkonjic, George Charames, Ken Kron, Jamie Rawson, Liyang Liu, Brian Li, Ted Chappell, Roula Raptis, Pinella

217

472. Mei M, Liu D, Dong S, Ingvarsson S, Goodfellow PJ, Chen H. The MLH1 -93 promoter

variant influences gene expression. Cancer Epidemiol;34(1):93-5.

473. Gazzoli I, Kolodner RD. Regulation of the human MSH6 gene by the Sp1 transcription

factor and alteration of promoter activity and expression by polymorphisms. Mol Cell Biol

2003;23(22):7992-8007.

474. Collins FS, Brooks LD, Chakravarti A. A DNA polymorphism discovery resource for

research on human genetic variation. Genome Res 1998;8(12):1229-31.